Melbourne, Australia - Cytokines

Transcription

Melbourne, Australia - Cytokines
Welcome
The International Cytokine and Interferon Society 2014 Annual Meeting
Cytokines Down Under: From Bench to Beyond
26th October – 29th October, 2014
Melbourne Convention & Exhibition Centre
Dear colleagues,
On behalf of the Scientific Organizing Committee, it is with great pleasure to welcome you to Melbourne to
attend the second annual meeting of the International Cytokine and Interferon Society (ICIS). Specific topics
will include the latest aspects on the biology, signal transduction and gene regulation of cytokines,
interferons and their receptors in innate and adaptive immunity, as well as pattern recognition receptors and
their role in host-pathogen interactions, infectious diseases, inflammation, cancer, autoimmunity and
metabolism. Sessions will include cutting edge basic science and clinical presentations in plenary and
concurrent symposia, as well as eminent keynote presentations, and are strongly supported by poster
sessions and trade displays.
The meeting promises to provide an outstanding forum for basic science and clinical researchers to present
their latest data and exchange ideas relating to the broad role of cytokines and interferons in human disease,
and applications to therapies. In addition, the meeting will provide strong networking opportunities for
scientists in the biotechnology and pharmaceutical industries. We are pleased with the attendance from all
over the globe by both established and new investigators and students –Thanks for your support. We thank
the Society and all sponsors who have helped to make this happen. This broad attendance, will help assure
a vibrant and exciting conference for all.
We also note that Australia, and Melbourne in particular, is a perfect location to visit at this time of year,
being in the peak of Spring. Melbourne is a vibrant and multicultural city known as the “sporting capital of
Australia”, being home to the Australian Tennis Open, Australian Formula 1 Grand Prix, and the Spring
Racing Carnival featuring the Melbourne Cup (first Tuesday in November), one of the largest and most
prestigious horse races in the world. Together with its renowned riverside atmosphere, café culture, nearby
beaches and wineries, it is no surprise Melbourne has been voted the most liveable city in the world for the
4th year in a row, with other Australian capital cities Adelaide, Sydney and Perth also featuring in the top 10.
We trust you will have a most productive conference and memorable experience whilst in Australia.
Brendan Jenkins and Paul Hertzog
Co-convenors
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 1
Index
Page
SECTION 1 – INTRODUCTION AND DELEGATE INFORMATION
Welcome
1
Contents
2
Organising Committee
3
Sponsors
4
Keynote Speakers
5
Symposia Speakers
6
2014 Award Winners
17
Delegate Information
18
Venue Map
21
Melbourne Information
22
Sponsor / Exhibitor Listing & Map
23
SECTION 2 – SCIENTIFIC PROGRAM
Full Program :
Sunday
28
Monday
30
Tuesday
35
Wednesday
39
SECTION 3 – PRESENTING AUTHOR INDEX
43
SECTION 4 – ABSTRACTS
Speaker Abstracts
Meeting Abstracts
SECTION 5 – DELEGATE LIST
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 2
67
70
138
Organising Committee
Convenors
Paul Hertzog, MIMR-PHI Institute of Medical Research,
Victoria, Australia
Brendan Jenkins, MIMR-PHI Institute of Medical
Research, Victoria, Australia
Secretariat
Maree Overall, ASN Events Pty Ltd
Kara Taglieri, ASN Events Pty Ltd
Local Organising Committee
Iain Campbell, University of Sydney, NSW, Australia
Angel Lopez, Centre for Cancer Biology, SA, Australia
Ashley Mansell, MIMR-PHI Institute of Medical Research,
Victoria, Australia
Sandra Nicholson, Walter and Eliza Hall Institute of
Medical Research, Victoria, Australia
Matt Sweet, Institute for Molecular Bioscience,
Queensland, Australia
International Scientific Advisory Committee
Vishva Dixit, Research Genentech, Inc., United States
Eleanor Fish, University Health Network, Canada
Sarah Gaffen, University of Pittsburgh, United States
Simon Jones, Cardiff University, United Kingdom
Doug Hilton, Walter and Eliza Hall Institute of Medical
Research, Australia
Hong Tang, Chinese Academy of Sciences, China
Luke O’Neill, Trinity College Dublin, Ireland
Masanobu Oshima, Kanzawa University, Japan.
Leon Platanias, Robert H. Lurie Comprehensive Cancer
Center, United States
Stefan Rose-John, University of Kiel, Germany
Bryan Williams, MIMR-PHI Institute of Medical Research,
Australia
Howard Young, National Cancer Institute at Frederick,
United States
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 3
Sponsors
The Organising Committee for the International Cytokine & Interferon Society 2014 Meeting acknowledges
with gratitude, the generous support received from the following sponsors:
GOLD SPONSORS
SILVER SPONSORS
BRONZE SPONSORS
SPONSORS
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 4
Keynote Speakers
Peter Doherty, University of Melbourne
Professor Peter Doherty shared the Nobel Prize in Physiology or Medicine in 1996
with Swiss colleague Rolf Zinkernagel, for their discovery of how the immune system
recognises virus-infected cells. He was Australian of the Year in 1997, and has since
been commuting between St Jude Children’s Research Hospital in Memphis and the
Department of Microbiology and Immunology at the University of Melbourne. His
research is mainly in the area of defence against viruses. He regularly devotes time to
delivering public lectures, writing articles for newspapers and magazines and
participating in radio discussions.
Peter Doherty graduated from the University of Queensland in Veterinary Science and
became a veterinary officer. Moving to Scotland, he received his PhD from the University of Edinburgh
Medical School. He is the first person with a veterinary qualification to win a Nobel Prize. Peter is also the
author of several books, including “A Light History of Hot Air”, “The Beginners Guide to Winning the Nobel
Prize” and "Sentinel Chickens: What birds tell us about our health and the world".
Nicos Nicola, Walter and Eliza Hall Institute of Medical Research
Dr. Nicola’s laboratory has focused on the molecular regulation of haemopoietic cell
production and function for over 30 years. A major emphasis has been on the
cytokines that regulate the production and functional activation of granulocytes and
macrophages, two types of white blood cells that co-ordinate innate immune
responses to bacterial and viral infections. His work has led to the identification,
purification and/or molecular cloning of several important cytokines (G-CSF, GM-CSF
and leukaemia inhibitory factor (LIF)), the identification and molecular cloning of
several cytokine receptors (including the GM-CSF, interleukin-11 and interleukin-13
receptors) and the identification of a new family of inducible, intracellular inhibitors of
these cytokine/receptor signaling pathways. These suppressors of cytokine signaling (SOCS) proteins have
been shown to have crucial roles in vivo in limiting the extent and duration of responses to various cytokines
thus preventing excessive inflammatory and autoimmune responses. He is also investigating the role of
cytokines and cytokine signaling pathways in the development and maintenance of leukaemic cell
populations and the usefulness of cytokines or cytokine antagonists as therapies or adjunct therapies in
cancer treatments.
Luke O’Neill, Trinity College Dublin
The major focus of the group is to provide a molecular understanding of innate
immunity and inflammation. We are interested in receptors involved in innate
immunity, such as Toll-like receptors (TLRs) and Nod-like receptors (NLRs – including
Nlrp3), and also signals activated, including NF-kappaB, IRF family transcription
factors and MAP kinases. The role played by this system in inflammatory diseases is
also under investigation. We have several projects underway including – Role of the
adapter Mal in the epithelial barrier in the gut, Control of TLRs and NLRs by
microRNAs, Role of Nlrp3 in Type 2 diabetes, Novel proteins in the TLR and NLR
systems, The role of Btk in TLR signalling, The control of trafficking of TLR4, Genetic
variation in innate immune genes and inflammatory diseases, Role of IL-36 in inflammation.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 5
Symposia Speakers
Shizuo Akira, Osaka University
Shizuo Akira is a director and professor of WPI Immunology Frontier Research
Center, and also a professor in Institute for Microbial Diseases at Osaka University,
Japan. He received his M.D. and Ph.D. from Osaka University. After two years of
postdoctoral working in Department of Immunology, University of California at
Berkeley, he started to study on IL-6 gene regulation and signaling in the Institute for
Molecular and Cellular Biology, Osaka University, and cloned transcription factors,
NF-IL6(C/EBP beta) and STAT3. He was a professor in Department of Biochemistry,
Hyogo College of Medicine from 1996 to 1999, where he became involved in Toll-like
receptors research. By generating TLR family knockout mice, he identified ligands of
many TLR members. He also demonstrated that the difference in signaling pathway among TLRs is due to
selective usage of adaptor molecules such as MyD88 and TRIF. He demonstrated that pathogen-derived
RNA is recognized by cytoplasmic receptor family, besides TLRs, and clarified the molecular mechanism of
antiviral response against RNA viruses. His current research interests are molecular mechanisms of innate
immunity and inflammation, which are studied mainly by generating knockout mice.
Awards and Honors: 2004 Robert Koch Prize (Robert Koch Foundation, Germany), 2006 William B. Coley
Award for Distinguished Research in Basic Immunology (Cancer Research Institute, USA), 2007 Imperial
Prize and Japan Academy Prize (Japan Academy), 2009 National Academy of Sciences of USA, Foreign
Associate, 2009 Person of Cultural Merit (Japanese Government), 2010 Keio International Medical Science
Prize (Keio University), 2010 Avery-Landsteiner Prize (German Society for Immunology), 2010 EMBO
Associate Member
Mark Ansel, University of California San Francisco
Lymphocyte lineage decisions are critical for the development of protective immunity
against a great diversity of pathogens, but improper or exaggerated responses also
contribute to the development and pathology of autoimmune diseases, chronic
inflammation, allergy, and asthma. The Ansel lab's primary experimental system is the
differentiation of the central coordinators of adaptive immune responses -- helper T
cells. Their distinct cellular identities (Th1, Th2, Th17, etc.) and associated functions
are defined by characteristic gene expression programs. We and many others have
documented how these programs are controlled by transcription factors, the cisregulatory DNA elements to which they bind, and epigenetic modifications that
constrain chromatin accessibility at those sites.
Most of our current work focuses microRNAs (miRNA) regulation of helper T cell behavior and immune
function, and how immunogenic stimuli regulate miRNAs homeostasis. Naive CD4+ T cells that cannot
produce any miRNAs exhibit reduced cell division and survival in response to immune stimuli. Surprisingly,
they also undergo rapid unrestrained differentiation into effector cells. One of the goals of our research is to
determine which specific miRNAs regulate each of these T cell behaviors, and which protein coding mRNAs
they target to exert their effects. In addition, we learned that T cells rapidly reset their miRNA repertoire upon
activation. This rapid change in miRNA expression may be important to allow T cells to change their gene
expression programs and develop effector functions.
Frances Balkwill, Barts Cancer Institute
My research is focused on the links between cancer and inflammation, being
especially interested in translating knowledge of cancer biology into new biological
treatments for cancer and in the role that inflammatory cytokines play in cancer
promotion.
My key areas of interest are: The Tumour Microenvironment, Inflammatory cytokines
and chemokines in the tumour microenvironment, 'Ovarian' cancer with a focus on
high grade serous and clear cell subtypes, Public engagement with biomedical
science and science policy.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 6
Jeff Babon, Walter and Eliza Hall Institute for Medical Research
Maintenance of the blood system is largely controlled by the secretion of small
glycoprotein messengers called cytokines. Cytokine exposure initiates an
intracellular signalling cascade that is driven by activation of a family of receptorbound tyrosine kinases known as JAKs (Janus Kinases). Under physiological
conditions JAK activation and signalling is tightly regulated, in particular, by the
SOCS (Suppressors of Cytokine Signalling) proteins.
However, in certain disease situations, mutant JAK overcomes its regulation by
SOCS, leading to haematological malignancies. One example of this is the
myeloproliferative disorders; a group of conditions that cause haematological cell
types (platelets, white blood cells, and red blood cells) to grow abnormally in the bone marrow. Our
laboratory uses the techniques of Structural Biology, Biochemistry and Biophysics to uncover the mechanism
of JAK/STAT signalling and its regulation by the SOCS proteins.
Gabrielle Belz, Walter and Eliza Hall Institute for Medical research
Our specific research interests include:
Deciphering the genetic control of effector and memory T cell differentiation,
Unravelling the transcriptional regulation of dendritic cell development and function,
Determining the control of innate immune cell subsets and their contribution to
protective immunity during infection, Transcriptional and molecular regulation of
peripheral lymphocyte differentiation
We are currently studying the following research areas: Development and
specialization of dendritic cells, Id and E protein regulation of effector and memory T
cell differentiation in infection, Transcriptional networks controlling T cell
differentiation, Development and function of innate immune cells.
Our research is focused on understanding how a subset of white blood cells, called cytotoxic T cells (CTL, or
CD8 T cells), recognise and remove virally-infected cells from the body following infection. The aim is to
identify specific factors that determine how virus-specific killer T cells develop during an infection and how
they impact on the establishment of immune memory after virus infection.
Our laboratory uses a combination of cutting-edge cellular and molecular technologies to dissect and study
virus-specific T cell responses to Influenza A and herpes viruses. These include techniques such as flow
cytometry, cell sorting, PCR and imaging.
Andrew Brooks, University of Queensland
Dr Andrew Brooks is a Senior Research Officer in the Institute for Molecular
Bioscience (University of Queensland, Australia) and joined the research group
headed by Prof Michael Waters in 2006. Andrew completed his Honours research on
Flaviviruses in 1996 at the Department of Microbiology and Immunology at James
Cook University and then moved to the Department of Biochemistry to study Dengue
Virus where he completed his PhD in 2002. He then moved to St Jude Children’s
Research Hospital in Memphis, TN, USA where he researched the role of EpsteinBarr Virus in oncogenesis. Andrew’s research interests are in cell signalling,
oncogenesis, and virology. His current research focus is on the molecular mechanism
of Growth Hormone mediated signalling via the Growth Hormone Receptor (GHR), the role of GHR in
oncogenesis, and the role of Growth Hormone signalling in liver regeneration. This research has led to
publications in journals including Science, Nature Cell Biology, PNAS, and Molecular Endocrinology. He has
been the recipient of over $1.5 million in research grant funding and has a number of national and
international collaborations. Andrew is an Editorial Board member for the Journal JAK-STAT and has been a
committee member of Australian Early-Mid Career Researchers Forum (AEMCRF) launched by the
Australian Academy of Science.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 7
Yinon Ben-Neriah, Hebrew University-Hadassah Medical School
Studying signaling pathways in animal models.
Cell growth and other processes associated with cancer are regulated by several
major signaling pathways, among which are the NF-kB, Wnt and the p53 activation
pathways, which are of major interests to us. We have elucidated some of the key
steps in NF-kB activation, including the degradation of the NF-kB inhibitor IkB (Alkalay
et al, PNAS 1995; Yaron et al, EMBO J, 1997 & Nature 1998) and identified CKIα as a
key component in the β-catenin destruction machinery at the core of the Wnt signaling
pathway (Amit et al, Genes & Development 2002, Elyada et al, Nature 2011). We
learned that studying signaling pathways in animal models is far more rewarding than tissue culture studies,
and a great deal of our lab work today is carried out in genetically engineered mouse models. We engineer
many model mice by ourselves, breed them onto different mutated mice and examine the models for the
purpose of identifying new disease mechanisms that explain complex human pathologies.
Much of our work it is done in collaboration with colleagues in Israel and abroad, Eli Pikarsky, Moshe Oren,
Varda Rotter, Aaron Ciechanover and Kari Alitalo.
Zhijian James Chen, University of Texas Southwestern Medical Center
We are broadly interested in mechanisms of signal transduction, namely how a cell
communicates with its surroundings and within itself. In particular, we are fascinated
by how a cell detects harmful or foreign insults and mounts an appropriate response
to restore homeostasis.
Our research revolves around the common theme of cell signaling and host defense:
Ubiquitin signaling in the NF-κB pathway, Innate immune sensing and signaling of
cytosolic nucleic acids.
Daniel Cua, Merck research Laboratories
Daniel Cua is the group leader in Pathways of Inflammation at Merck Research
Laboratories in Palo Alto and was a previous Research Fellow at Schering Plough
Biopharma. His work focuses on the discovery of novel biologic agents for the
treatment of immune-mediated inflammatory diseases.
Since 2003, Cua also has contributed to scientific and medical publications, with
more than 60 original articles published in prestigious journals such as Nature, Cell,
Immunology, Journal of Experimental Medicine and Journal of Clinical Investigation.
In addition, he is the co-inventor on 21 granted patents and applications.
Vojo Deretic, University of New Mexico
Vojo Deretic's main contributions to science come from studies by his team on the
role of autophagy in infection and immunity. Autophagy, a cytoplasmic pathway for
the removal of damaged or surplus organelles, has been previously implicated in
cancer, neurodegeneration, development, and aging. Dr. Deretic's group is one of
those that made the discovery that autophagic degradation is a major effector and
aregulator of innate and adaptive immunity mechanisms for direct elimination of
intracellular microbes (such as Mycobacterium tuberculosis). His current work is on
the role of autophagy in immunity and inflammation.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 8
Mark Febbraio, Baker Institute
Professor Mark Febbraio is a Senior Principal Research Fellow of the NHMRC, is the
Head of the Cellular and Molecular Metabolism Laboratory and Head of the Division
of Signalling & Metabolism at the Baker IDI Heart & Diabetes Institute. He is also the
Chief Scientific Officer and on the Board of Directors of N-Gene Research
Laboratories Inc., a USA based Biotechnology company. His research is focussed on
understanding cellular and molecular mechanisms associated obesity and type 2
diabetes. He has authored over 200 peer reviewed papers in leading journals such as
Nature, Nature Medicine, Cell, Cell Metabolism, Nature Immunology, The Journal of
Clinical Investigation, PNAS and Diabetes. He has an H factor of 64 and over 11,000
career citations. He has won prizes at international, national and institutional levels including the A K
McIntyre Prize for significant contributions to Australian Physiological Science (1999), the Colin I Johnson
Lectureship by the High Blood Pressure Research Council of Australia (2006) the ESA/ADS Joint Plenary
Lecture (2009) and the Sandford Skinner Oration (2011). He is on the Editorial Board of Diabetes, The
American Journal of Physiology Endocrinology & Metabolism, and the Journal of Applied Physiology.
Professor Febbraio is also dedicated to health and fitness and continues to complete in running races and
multi-sport events.
Richard Flavell, Yale University School of Medicine
Richard Flavell is co-discoverer of introns in cellular genes: he showed DNA
methylation correlates inversely with, and prevents, gene expression. He was the first
to develop reverse genetics as a postdoc with Weissmann and in his own lab
continued in this field throughout his career; he is a pioneer in the use of this approach
in vivo to study function. Dr. Flavell’s laboratory studies the molecular and cellular
basis of the immune response. He has been instrumental in discovering the molecular
basis of T-cell differentiation from precursor cells into differentiated subsets. This work
led to the discovery of GATA3 as a critical regulator of the Th2 response and the first
example of such a molecule in Th cell differentiation. He went on to demonstrate the
first case of regulation of gene expression in trans, via ”chromosome kissing.” Moreover his laboratory has
elucidated the mechanisms of immunoregulation which prevent autoimmunity and overaggressive responses
to pathogens. Specifically, Dr. Flavell's laboratory has elucidated the role of TGF-β in the regulation of
immune response. This work is of relevance both to the control of autoimmune disease and the evasion of
immune response by tumors.
Dr. Flavell’s laboratory has discovered the role of several receptor families in the innate immune response,
including the role of several Toll-like receptors and intracellular Nod-like receptor families (NLRs). This has
recently led to the elucidation of function of Nod2 in inflammatory bowel diseases and Nlrp proteins in the
production of IL-1. Most recently he has established a fascinating connection between inflammasomes,
microbial homeostasis and chronic diseases. He showed that inflammasome dysfunction causes dysbiosis of
the microbiota which, in conjunction with a susceptible diet, leads to IBD and Metabolic Syndrome, including
Obesity, Fatty Liver disease and Type 2 diabetes.
Frederic Geissman, King’s College
The CMCBI was recently established in a new laboratory space in King's College
London (Division of Immunology, Infection and Inflammatory Diseases) with the
support of Arthritis Research UK.
We aim to advance knowledge of the molecular and cellular mechanisms of
inflammation, and to open roads to innovative treatment of inflammation and
inflammatory diseases.
We use both molecular approaches and in vivo model systems, to investigate the
molecular and cellular pathways and networks that control inflammation. Research
teams work on basic models of inflammation, as well as on human diseases.
Research teams develop extensive collaborations among themselves and with other groups in the DIIID, the
Randall Division and with Researchers across the world. Tools available in the lab include intravital
microscopy, flow cytometry and cell sorting, mouse husbandry, and a Fly lab.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 9
Florent Ginhoux, Agency for Science, Technology and Research
(A*STAR)
My laboratory focuses on the biology of dendritic cells, which are very unique and
special cells of the immune system. They are crucial pathogen sensing and antigen
presenting cells that basically control the initiation of our body immune responses to
any invading microbes or vaccines that we receive. We are trying to understand
where do these cells come from in both mice and humans, how do they work and
how they are made in order to better manipulate them in vitro and in vivo.
Elizabeth Hartland, University of Melbourne
Elizabeth Hartland obtained her B.Sc. (Hons) majoring in Microbiology and
Biochemistry and subsequently her Ph.D in Microbiology from the University of
Melbourne. She has held a Royal Society/NHMRC Howard Florey Fellowship in the
Department of Biochemistry, Imperial College London, Lecturer/Senior Lecturer
positions in the Department of Microbiology, Monash University, Australia and an
Australian Research Council Future Fellowship at the University of Melbourne. She
is currently Professor of Microbiology and Head of the Department of Microbiology
and Immunology at the University of Melbourne. Professor Hartland has a longstanding research interest in the pathogenesis of infections caused by Escherichia
coli and Legionella, with a focus on mechanisms of bacterial colonization and immune evasion.
Veit Hornung, University of Bonn
In our research projects, we are trying to understand what mechanisms are
employed by our innate immune system to distinguish self from non-self. Central to
this complex task is a repertoire of pattern recognition receptors (PRRs) that have
evolved to detect the presence of microorganisms. The ligands or targets of these
PRRs are usually referred to as pathogen-associated molecular patterns (PAMPs).
PAMPs are typically molecular structures that are unique to the physiological
processes of microorganisms but not the host. In addition, PAMPs usually
constitute products that are essential to microorganisms and thus cannot be
changed or lost by the respective microorganism via adaptive evolution. In addition
to the recognition of PAMPs, some PRRs can also detect endogenous danger and
stress situations. Cell stress or tissue damage can lead to the release of normally compartmentalized
molecules or the chemical modification of self-molecules. These and other endogenous inflammatory signals
that appear after cellular damage or due to metabolic derangements are collectively known as dangerassociated molecular patterns (DAMPs), and their ability to trigger inflammation is mediated by the PRRs of
our innate immune system.
Zhengfan Jiang, Peking University
Innate immunity is the first line of defense against microbial invasions via a set of
pattern-recognition molecules. We have established a powerful cDNA library
screening system to search systematically for new genes essential in innate
immunity. Using Yeast two hybrid screen, immuno-purification and Mass-spec-based
protein identification techniques, we are also interested in identifying new genes in
the signal complexes. Current projects in the lab focus on 1) Mechanism through
which cells recognize dsDNA. 2) Function of ubiquitination in innate immunity. 3)
Mechanisms leading to cytokine production and secretion after infection. 4)
Malfunction of innate immunity and related diseases.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 10
Simon Jones, Cardiff University
It is likely that transition from the recruitment of leukocytes typically associated with
innate immunity to those important in acquired immunity facilitates resolution of
inflammation, and the restoration of normal tissue architecture. These findings
emphasise a central role for IL-6 in the regulation of this step, and as such may be
important in facilitating successful resolution of acute inflammation. Paradoxically, IL-6
concentrations are increasingly being monitored as a clinical marker of severity during
ongoing inflammation or as an indicator of disease outcome. Our studies illustrate that
in more chronic conditions like rheumatoid arthritis, sIL-6R-signalling elicits more
detrimental consequences, and we have found that blockade of sIL-6R activity using
soluble gp130 (sgp130) as a natural sIL-6R antagonist suppresses development of antigen-induced arthritis
[Nowell et al., 2003 Journal of Immunology 171:3202; Richards et al., 2006 Arthritis & Rheumatism, In
press]. Consequently IL-6 responses depend either on the inflammatory condition or its activities become
dysregulated during more chronic inflammatory episodes. Ultimately these questions form the rationale basis
for future investigations. In particular, translational studies in association with Professor Nicholas Topley are
currently exploring the novel concept of ‘resolution therapeutic®’. This therapeutic strategy is based on
supplementing host defence to re-synchronise the immune response and in so doing promote inflammatory
resolution. With Wellcome Trust support we have developed an IL-6-sIL-6R chimeric fusion protein, which
we propose will aid resolution of local bacterial infections by facilitating transition from innate to acquired
immunity.
Thirumula-Devi Kanneganti, St Jude Children’s Research Hospital
Our research aims to understand how the innate immune system recognize and
respond to pathogens and how mutations in these sensing systems affect the
development of infectious, inflammatory, and autoimmune diseases in humans. Our
initial studies examined the roles of TLRs, NLRs and their adaptor proteins in
response to certain microbial components (Nature 440(7081):233-36; Nature
Immunology 7(6):576-82; JBC 281(48):36560-68; Journal of Immunology
178(12):8022-27; Immunity 26(4):433-43). Ongoing studies are investigating the
sophisticated system governing activation of signaling cascades and the mechanisms
involved (Mol Cell Proteomics 7(12):2350-63; Blood 113(12):2742-5; Immunity
32(3):379-91).
Eicke Latz, University of Bonn
The innate immune system responds to microbial products and host factors that arise
during tissue damage and metabolic dysregulation. Innate immune activation is
important for the control of infections and overactivation can lead to inflammatory
disease states. An international group of scientists work together with the major aims
of: elucidating how the innate immune system maintains health and under which
circumstances it promotes diseases, using basic research to decipher the molecular
mechanisms of innate immune activation, developing novel therapeutic approaches
that target a multitude of inflammatory diseases, such as Alzheimer's disease,
diabetes, and atherosclerosis.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 11
Fabienne MacKay, Monash University
Prof. Mackay’s laboratory has an interest in autoimmune diseases and mechanisms
leading to loss of immune tolerance, in particular that of B-lymphocytes. Prof
Mackay has spent years studying a cytokine from the TNF superfamily named
BAFF/BLyS and demonstrated the role of this factor in B cell survival. Excess BAFF
leads to autoimmunity in mice and is associated with human autoimmunity, in
particular systemic lupus erythematosus (SLE) and Sjögren’s syndrome. Inhibitors
of BAFF have recently met the primary endpoints in a phase III clinical trial with SLE
patients, validating in human patients the critical role of the BAFF system in SLE.
BAFF drives autoimmunity independently of T cells but this process requires the
function of Toll-like receptors (TLR) and as such drives an unusual form of autoimmunity. BAFF signals
through three receptors BAFF-R, TACI and BCMA. BAFF-R triggers survival but is also express on activated
T cells and regulatory T cells on which the role of BAFF-R is still obscure. TACI is expressed on a subset of
B cells, monocytes and dendritic cells, and its expression is regulated by TLR activation. TACI is required for
T-independent antibody responses but also negatively regulate B cells. Its role as a negative regulator of B
cells remains unclear. TACI appears to be activated by oligomeric forms of BAFF and APRIL crosslinked by
Heparan Sulfate Proteoglycans (HSPG) but not by trimeric ligands. BCMA is expressed on plasma cells and
is required for cell survival. The function of APRIL another ligand for TACI and BCMA versus BAFF is also
not fully understood. APRIL is highly expressed by cancer cells and it may drive TACI-dependent immune
mechanisms in tumors. All these issues are currently addressed in Prof. Mackay’s laboratory.
Prof. Mackay’s lab also works on the chemokine receptor CXCR7, which is important for heart formation and
tissue remodelling. CXCR7 deficiency delays some autoimmune conditions in mouse models and its role in
T cells will be addressed.
Finally, Prof Mackay has worked on the role of neuropeptide Y (released in response to psychological stress)
and its receptors in suppressing immune functions and protection against cancers.
Kingston Mills, Trinity College
Kingston Mills is Professor of Experimental Immunology, School of Biochemistry and
Immunology, Trinity College Dublin (TCD). He is Head of The Centre for the Study of
Immunology at Trinity Biomedical Sciences Institute and Theme Champion for
Immunology, Inflammation and Infection at TCD. He is a graduate of TCD and
trained at as a Postdoctoral Fellow at University College London and the National
Institute for Medical Research, Mill Hill, London, before joining the Scientific Staff of
NIBSC, Herts, UK. He returned to Ireland in 1993 to take up an academic position at
National University of Ireland, Maynooth. He was appointed to a Personal Chair at
Trinity College Dublin in 2001 and was Head of the School of Biochemistry and
Immunology from 2008-2011. He heads an active research team focusing on T cells in infection,
autoimmunity and cancer. He is co-founder of Opsona Therapeutics and TriMod Therapeutics, biotech
companies focusing on the development of immunotherapeutics for inflammatory diseases and cancer.
Denise Monack, Stanford University
The primary focus of our research is to understand the genetic and molecular
mechanisms of intracellular bacterial pathogenesis. We use two model systems,
Salmonella typhimurium and Francisella tularensis, to study the complex hostpathogen interactions.
Both of these organisms survive and multiply in macrophages, an important immune
effector cell. Macrophages express Pattern Recognition Receptors on the surface as
well as in the cytosol. Our laboratory focuses on the cytosolic recognition of bacteria
that leads to Type I Interferon signaling and Inflammasome activation. We take both a
genetic and biochemical approach to understand the molecular mechanisms involved
in host recognition pathways leading to inflammation and pathogen evasion mechanisms.
Salmonella typhi causes the systemic disease typhoid fever and Francisella tularensis causes the systemic
disease tularemia (“rabbit fever”). Utilizing mouse models of systemic salmonellosis and tularemia, we would
like to understand how Salmonella persists within certain hosts for years in the face of a robust immune
response and how F. tularensis, a stealth invader, can cause a rapid, lethal infection.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 12
Masaaki Murakami, Osaka University
Professor, Division of Molecular Neuroimmunology, Institute for Genomic Medicine
and Graduate School of Medicine, Hokkaido University.
Education: 1983-1989 Hokkaido University, School of Veterinary Medicine, 1989-1993
Osaka University, Graduate School of Medical.
Research and career: 1993-1999 Assistant Professor, Institute of Immunological
Science, Hokkaido University, 1999-2001 JST fellow, HHMI, National Jewish Medical
and Research Center, 2001-2003 Visiting Associate Professor, Colorado University,
Integrated Department of Immunology, 2002-2013 Associate Professor, Graduate
School of Medicine and Frontier Biosciences, Osaka University, 2014-present
Professor, Institute for Genomic Medicine and Graduate School of Medicine, Hokkaido University.
Research field:Neuro-immune interaction for the development of inflammation
Gabriel Nunez, University of Michigan
The Nuñez laboratory is interested in signaling pathways regulating innate immunity,
the pathogenesis of inflammatory disease and cancer. Specifically, the research
focuses on mechanistic studies to understand the role of proteins of the Nod-like
receptor (NLR) and Toll-like receptor (TLR) families in the host immune response
against microbial pathogens and endogenous danger signals. Several approaches
that include analyses of genetically modified mutant mice, cell biology and
biochemical studies are used to determine molecular mechanisms involved in the
interaction between microbial/endogenous molecules and innate immune receptors.
Several NLR proteins including Nod2 and Nlrp3/Nalp3 are mutated in patients with
inflammatory diseases (Crohn's disease and autoinflammatory syndromes). Furthermore genetic variationin
NLRs affect the susceptibility to asthma and sarcoidosis. Studies to understand how the NLR mutant
proteins lead to disease are a major effort of the laboratory. Signaling pathways involved in inflammation and
innate immunity are also known to play a role in cancer development particularly in the gastrointestinal tract.
Understanding the mechanisms by which these pro-inflammatory pathways affect cancer initiation and/or
progression in the intestinal tract is another interest of the laboratory.
Belinda Parker, La Trobe University
Our laboratory has a particular focus on determining the mechanisms of tumour cell
spread to distant tissues and organs. In cancers such as breast and prostate, spread
to distant organs can occur years after primary tumour removal and the treatments
available for cancer patients with metastatic disease in tissues such as bone are
rarely curative. For this reason, the focus of the Cancer Microenvironment and
Immunology group is to determine the properties of tumour cells and interacting cells
in the surrounding tissue that promote metastatic spread in clinically relevant models
of breast and other cancers. Our ultimate goal is to predict patients that are likely to
develop distant spread and to design new anti-metastatic therapies that block the
invasion and growth of cancer cells in these distant tissues. Most models used to study bone metastasis
involve injection of human cells into the blood stream of immunocompromised mice and do not allow
interaction between tumour cells and stromal cells (other cells that interact with cancer cells in a tumour), nor
permit analysis of the contribution of the immune system to metastasis. Using a model of breast cancer that
mimics breast cancer metastasis and allows appropriate interaction between stromal cells and the immune
system, our group has identified two pathways that are important in bone metastasis: A group of proteases
called the cysteine cathepsins and their inhibitors that have an important role in tumour cells and surrounding
stromal cells in invasion of breast cancer cells at the primary site and in metastatic tissues.
An immune defence pathway called the Type I IFN pathway that is produced by cancer cells and stimulates
an immune response similar to the signals produced by infection. We have found that cancer cells suppress
these signals to spread to the bone and that restoring the immune response has therapeutic value in breast
cancer models.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 13
Stefan Rose-John, University of Kiel
Since 25 years, my laboratory is focused on understanding the molecular biology of
cytokines. Using structure-function analysis approaches, we have in the past defined
the binding sites of Interleukin-6 and its two receptor subunits, Interleukin-6-receptor
and gp130. This has enabled us to build the first molecular model of the Interleukin-6
receptor complex and was the starting point for the construction of various designer
cytokines, which have considerably higher specific biologic activity than the natural
cytokines. A second aspect of our work has started with the discovery of a naturally
occurring proteolytic cleavage of the Interleukin-6 receptor, which leads to the
generation of a soluble Interleukin-6 receptor. We have discovered that the complex
of soluble Interleukin-6-receptor and Interleukin-6 stimulates cells, which express the signal transducing
receptor subunit gp130 but not the ligand binding subunit Interleukin-6 receptor. In the absence of the sIL-6R
such cells do not respond to IL-6. We have called this process 'transsignaling'.
We have shown in the past 10 years that 'trans-signaling' is important for the regulation of cellular
differentiation and of apoptosis and has a prominent role in inflammation, neuronal survival, hematopoiesis,
and tumor defense. We are currently developing the concept that the triggering of 'transsignaling' is an
emergency reaction of the human immune system and that disruption of 'trans-signaling' can be used
therapeutically for the treatment of chronic inflammatory diseases and cancer. Indeed, one cytokine
antagonist, which – in animal models – has proven effective in blocking chronic inflammatory diseases such
as Crohn’s diseases, Rheumatoid Arthritis and inflammatory colon cancer, is now tested in Phase I clinic
trials since June 2013.
Chuck Samuel, University of California Santa Barbara
The overall objective of the research in The Samuel Lab is to elucidate in molecular
terms the mechanisms by which interferons exert their antiviral and cell growth control
actions in mammalian cells. Present work includes biochemical and molecular genetic
studies of two interferon-inducible enzymes, PKR and ADAR. PKR is a doublestranded RNA-dependent protein kinase induced by IFN, and activated by RNAdependent autophosphorylation. PKR plays a major role in the regulation of translation
of viral and cellular mRNAs and also modulates transcription and signaling. The
ADAR1 deaminase is an RNA editing-enzyme that catalyzes the C-6 deamination of
adenosine to yield inosine, thereby altering the genetic decoding and structure of
RNAs. While PKR displays antiviral and proapoptic activities, ADAR1 is often proviral and antiapoptotic in
virus-infected cells. Furthermore, PKR is not required for normal mouse embryogenesis, whereas ADAR1 is
required.
Feng Shao, National Institute of Biological Sciences
Dr. Feng Shao is an investigator at National Institute of Biological Sciences (NIBS),
Beijing, CHINA. He was a chemistry undergraduate of Peking University from 1991 to
1996 and received his master degree from Institute of Biophysics, Chinese Academy
of Sciences in 1999. Dr. Shao was trained as a biochemist and obtained his PhD
degree with Dr. Jack E. Dixon from University of Michigan in 2003. Prior to returning
to China in 2005 to assume an assistant investigator and group leader position at
NIBS, he was a Damon Runyon Postdoc Research Fellow at Harvard Medical
School. Dr. Shao was promoted to be an associate investigator in 2009 and a full
investigator in 2012 at NIBS.
Dr. Shao’s research focuses on biochemical
mechanism of bacterial infection and host innate immune defense. His laboratory has
discovered several novel post-translational modifications used by the bacteria to disrupt host immune
defense as well as innate immune sensors for various bacterial molecules and toxins. Dr. Shao has authored
40 research articles in highly-regarded journals, including 10 in Nature, Science and Cell, and his work has
been recognized by several renowned academic honors including the International Early Career Award from
Howard Hughes Medical Institute and the Irving Sigal Young Investigator Award from the Protein Society. Dr.
Shao also serves as journal editors for eLife and Cellular Microbiology.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 14
Mark Smyth, Queensland Institute of Medical research
Professor Mark Smyth is regarded as an international leader in tumour
immunology/immunotherapy and natural killer (NK) cell biology. He received his PhD
from the University of Melbourne in 1988 and trained at the National Cancer Institute
(USA) from 1988-1992. After 8 years at the Austin Research Institute in Melbourne,
working on mechanisms of lymphocyte-mediated cytotoxicity, he relocated to the
Peter Mac where his studies on effector molecules collectively rekindled world-wide
interest in cancer immune surveillance. In recent years, Professor Smyth has: made
key discoveries linking NK cells and adaptive T cell control of tumours; demonstrated
a unique combination of pathways that can potently reject established cancers in
mice; first defined immune-mediated dormancy of cancer; defined the role of host in chemotherapy
responses; and defined adenosine and CD96 as new targets for cancer immunotherapy. A number of his
discoveries have led to new clinical trials in cancer immunotherapy. In 2013, Professor Smyth relocated to
the QIMR Berghofer in Brsibane as a Senior Scientist and NH&MRC Australia Fellow. He is a Senior Editor
at Cancer Research, and a member of the Scientific Advisory Board of the Cancer Research Institute (USA).
Dale Umetsu, Genentech
Our lab focuses on the study of subpopulations of human and murine CD4+ T cells,
which play a central role in the regulation of adaptive immunity and tolerance. The
laboratory studies CD4+ T cells in humans and in mice, examining the function of
CD4+ alphaß TCR T cells, antigen-specific regulatory T cells, as well as iNKT cells in
regulating immune responses. We are interested in the cellular, molecular and
genetic mechanisms that control the interaction of T cells with dendritic cells and that
regulate cytokine synthesis in and the function of regulatory T cells and iNKT cells in
the respiratory and gastrointestinal tracts. We are particularly interested in the
function and immunology of the TIM family of genes, which we discovered several
years ago, and which regulate the development of autoimmune and allergic diseases.
Uwe Vinkemeier, University of Nottingham
1992 - PhD in Biochemistry, Max-Planck-Institut für Biophysikalische Chemie in
Göttingen; 1993-1999 - Post-doctoral Associate with James Darnell and John
Kuriyan at the Rockefeller University in New York; 2000-2007 - Head of Junior
Research Laboratory at the Leibniz-Institut für Molekulare Pharmakologie in Berlin;
2007 to date - Action Medical Research Chair of Cell Biology at The University of
Nottingham.
Xiao-Fan Wang, Duke University
Xiao-Fan Wang was born in Wulumuqi, China. He entered Wuhan University in 1978
to receive his college education following the reform of the education system. In 1982,
as one of the first group of Chinese students sent to study biology in the US, he
started his graduate training in transcriptional regulation of immunoglobulin genes
during B cell development with Dr. K. Calame at UCLA and received his Ph.D. in
1986. He then spent five years at Whitehead Institute and MIT as a postdoctoral
fellow under the guidance of Dr. R. Weinberg. The main achievement during this
period was the molecular cloning of transforming growth factor ß (TGF-ß) type II and
type III receptors. In early 1992 he moved to Duke University Medical Center as an
Assistant Professor in the Department of Pharmacology & Cancer Biology. He is currently Professor of
Pharmacology and Cancer Biology, Donald and Elizabeth Cooke Professor of Experimental Oncology. His
other academic activities include serving on the editorial boards of a number of scientific journals, such as an
Associate Editor for the Journal of Biological Chemistry. The current research in the Wang laboratory mainly
focuses on the elucidation of molecular nature and signaling mechanisms associated with tumor
microenvironment that promotes tumor progression and metastasis. Particularly, the lab is interested in the
roles of specific microRNAs as mediators of TGF-ß or hypoxia to affect the biological nature of tumor
microenvironment, via the interactions with the immune system and the recruitment of various types of
stromal cells, to enhance tumor metastasis. The lab is also studying the mechanisms underlying the
phenomenon of cellular senescence.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 15
Wolfgang Weninger, Centenary Institute
The laboratory is investigating how the cells of the innate immune system, the first
responders, interact with pathogens in skin tissue. These researchers are challenging
the tissue with different viruses, bacteria and parasites to test a variety of different
models of infection. The studies have implications for the development of vaccines.
Hua Yu, Beckman research Institute
Dr. Hua Yu is Co-Leader of Cancer Immunotherapeutics Program, City of Hope
Comprehensive Cancer Center and Associate Chair of the Department of Cancer
Immunotherapeutics and Tumor Immunology. Dr. Yu is a noted expert and pioneer on
the cancer-promoting protein STAT3 and was the first to uncover and define the
protein’s effect on the immune system. Dr. Yu’s studies have laid the foundation for a
new generation of molecular targeted cancer therapy approaches that disable both
tumor cells and the tumor stromal cells, which are critical for tumor growth. She has
developed a potentially paradigm-shifting new drug targeting STAT3 that will be
among the first produced in City of Hope’s new Chemical GMP Synthesis Facility.
Dr. Yu received her bachelor’s and doctoral degrees from Columbia University. She
completed fellowships with the American Cancer Society and the National Institutes of Health, and also was
a faculty fellow at Columbia University. The fundamental discoveries from her laboratory have been well
supported continuously by grants from the National Institutes of Health. Her recent studies have been
published extensively in such prestigious biomedical/cancer research journals as Nature Medicine, Cancer
Cell, Nature Biotechnology and Nature Reviews Cancer and Nature Reviews.
Elina I Zuniga, University of California San Diego
Viral persistence requires a long-term relationship between the host and the
microbe that involves multiple layers of interactions from molecular to cellular to
whole organisms. Our laboratory studies cellular and molecular aspects of virushost interactions during acute versus chronic viral infections to determine general
principles of viral immune-evasion, persistence and pathogenesis. The ultimate
goal is to modulate or recover immune system functions to prevent or eradicate
chronic viral infections. During the past years at UCSD, I have congregated a
group of young scientists with diverse expertise and strong motivation to obtain
solid answers for understanding immune responses during chronic viral infections,
a topic of major biomedical relevance.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 16
2014 Award Winners
Seymour and Vivian Milstein Award for Excellence in Interferon and Cytokine Research
Dr. Stefanie N. Vogel, University of Maryland School of Medicine, Baltimore, MD
Dr. Katherine A. Fitzgerald, University of Massachusetts Medical School, Worcester, MA
Honorary Lifetime Membership Award
Charles E. Samuel, University of California, Santa Barbara, Santa Barbara, CA
Amanda Proudfoot,
Milstein Young Investigator Award
Dr Dominic De Nardo, University of Bonn
Stacy Horner, Duke University Medical Center, Durham, NC
Maria Liaskos, MIMR-PHI Institute of Medical Research, Melbourne, Australia
Seth Masters, The Walter and Eliza Hall Institute, Victoria, Australia
Kate Schroder, The University of Queensland, Brisbane, Australia
Christina Fleischmann Award to Young Women Investigators
Sophie Broughton, St Vincents Institute For Medical Research, Victoria, Australia
ICIS Journal of Biological Chemistry / Herbert Tabor Award
Yeonseok Chung, Seoul National University, Seoul, South Korea
ICIS Sidney and Joan Pestka Award
Annie Bruns, Northwestern University, Evanston, IL
Postgraduate Award
Amanda Huber, University of Michigan, Ann Arbor, MI
Distinguished Service Award
Dr. Christine Czarniecki
ICIS & Melbourne Travel Award Winners
Ryann Guayasamin
Daniel Harari
Jeremy Hirota
Alan Hsu
Caleb Huang
Ian Humphreys
Iryna Kolosenko
Pankaj Kothavade
Andrew Larner
David Levy
Helene Minyi Liu
Yiliu Liu
Yueh-Ming Loo
Pei Ching (Regine) Low
Tao Lu
Pilvi Maliniemi
Isabelle Marie
Kazuya asuda
Mariko Matsui
Nontobeko Mvubu
Marcel Nold
Roza Nurieva
Olusegun Onabajo
Matthias Parrini
Hye-Lim Park
Sowmya Pattabhi
Leesa Pennell
Aradhana Rani
Mahesh Raundhal
Rebecca Coll
Nancy Reich
Fabiana Rizzo
Saleela Ruwanpura
Maria Sanchez-Aparicio
Marvin Sandoval
Karina Santiago
Smriti Sharma
Malcolm Starkey
Jason Twhohig
Lauren Vaughn
Nàdia Villacampa
Rui DanXie
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 17
Delegate Information
MEETING DATES
26 – 29 October, 2014
WEBSITE
www.cytokines2014.com
SECRETARIAT
ASN Events Pty Ltd
Unit 9
397 Smith Street
Fitzroy VIC 3065
Ph:
+61 3 8658 9530
Fax:
+61 3 8658 9531
VENUE
Melbourne Convention and Exhibition Centre
1 Convention Centre Place
South Wharf
VIC Australia 3006
Ph: +61 3 9235 8000
ORGANISER’S REGISTRATION DESK
The organiser’s registration desk will be located in the main foyer. The desk will be attended at all times
during the conference, see hours below. Delegates should collect their satchel, name tag and other
conference material on arrival. A message board will be at the registration desk.
ON SITE REGISTRATION DESK CONTACT HOURS
During open hours of event only:
th
Sunday 26 :
14:00 – 18:30
th
Monday 27 :
07:30 – 18:30
th
Tuesday 28 :
07:30 – 18:30
th
Wednesday 29 :
07:00 – 18:30
REGISTRATION
Conference delegates receive the following as part of their registration:
 Access to all scientific sessions and the exhibition hall on day(s) of registration
 Sunday Welcome Function;
 Morning teas;
 Lunches;
 Afternoon teas;
 Poster & network drinks
 A satchel with a copy of the delegate handbook and abstracts
*Not applicable for trade delegates unless supply allows
Also included on the days of registration are:
 Lunches (Monday, Tuesday, Wednesday)
 Morning teas (Monday, Tuesday, Wednesday)
 Afternoon teas (Monday, Tuesday, Wednesday)
 The Welcome Function on Sunday evening
 Drinks during the poster sessions 6:00 – 7:30pm (Monday and Tuesday nights)
NAME TAGS
The Congress entrance will be manned by security and delegates are required to wear their name tags to all
scientific and catered sessions.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 18
INTERNET
Melbourne Convention and Exhibition Centre have access to free wireless internet. Please select the ‘M
Connect’ wireless service on your device. Then, after opening your preferred internet browser, the M
Connect page will appear for you to read the terms and conditions and confirm your connection by selecting
‘Connect Now’.
ACCOMMODATION
Delegates are staying at a variety of hotels in the area. Most delegates are accommodated in the immediate
vicinity of the Convention Centre. Standard hotel check in is 2:00pm and check out is 10:00am. Delegates
seeking to vary this arrangement should contact the Hotel reception directly. Delegates are to settle their
own accommodation account directly with the hotel upon departure.
Crowne Plaza Melbourne
1-5 Spencer Street, Melbourne
+61 3 9648 2777
Rendezvous Grand Hotel Melbourne
328 Flinders Street, Melbourne
+61 3 9250 1888
Hilton Melbourne South Wharf
2 Convention Centre Place, Melbourne
+61 3 9027 2000
Melbourne Short Stay – MP Deluxe
Corner of City Road & Moray Street
+61 3 8256 7500
Melbourne Short Stay – Southbank Deluxe
63 Whiteman Street, Melbourne
+61 3 8256 7500
Vibe Hotel Savoy Melbourne
630 Little Collins Street, Melbourne
+61 3 9622 8888
Holiday Inn Melbourne on Flinders
575 Flinders Lane, Melbourne
+61 3 9629 4111
INSURANCE
The hosts and organisers are not responsible for personal accidents, any travel costs, or the loss of private
property and will not be liable for any claims. Delegates requiring insurance should make their own
arrangements.
DISCLAIMER
The hosts, organisers, venue and participating societies are not responsible for, or represented by, the
opinions expressed by participants in either the sessions or their written abstracts. Responsibility for the
literary and scientific content of abstracts accepted for publication remains with the authors and their
sponsoring institutions. Acceptance by any of the participating societies for publication does not imply any
acceptance by the Societies of responsibility.
SESSION LOCATIONS
The Plenary room is located on the Main Floor of the Convention Centre – near the registration desk. Some
catering will be provided here. Concurrent Symposia room is located on Level 1. Also located on Level 1 is
the trade hall, posters and more catering.
THE SPEAKER PREPARATION ROOM
The speaker preparation room is located on level 1 and is Speaker Room 101. As the program is large and
complex, it is essential you load your talk to the conference network from the speaker preparation room
(noted above) at least 2 hours in advance of your session start time. There will not be provision for people to
use their own laptops. The conference presentation software is MS PowerPoint 2007. Those preparing on
Macs should save for this output. Any issues should be resolved in the speaker preparation room beforehand
and there will be a technician present to assist.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 19
POSTER VIEWING
Delegates with posters can place their poster by finding the appropriate abstract number on the display
panels in the exhibition area. The program provides instructions regarding the days which posters should be
displayed and manned. Delegates with poster discussions on a particular day are encouraged be present at
their poster during the breaks on that day to answer questions and meet colleagues with similar research
interests. During the specific poster discussion sessions, presenters must be present talk discuss with
delegates.
SMOKING
Smoking is not permitted in the venue.
MOBILE PHONES
Please ensure your mobile phone is turned off during any session you attend.
SOCIAL FUNCTIONS
th
 Welcome Reception: Sunday 26 October, 7:30 - 9:00PM, Ground Level Foyer
Delegates are invited to the Welcome Function following the opening symposium on the evening of Sunday
th
October 26 , 7:30 – 9:00 PM. The registration desk will be open from 2:00pm. The opening formalities will
be included and this function is included with full registration.
th
th
 Poster Sessions: Monday 27 & Tuesday 28 October, 6:00 - 7:30 PM, Level 1 Foyer
Each of these evenings, delegates are invited to view posters and socialise with the trade participants and
other delegates for an hour at the end of the sessions. This is included in the registration of the day.
th
 Networking session for students and ECR’s:
Monday 27 October, 7:30PM, The
Boatbuilders Yard
Optional social event for students and ECR’s. Attendance includes 2 drink vouchers and nibbles.
th
 Networking Dinner: Tuesday 28 October, 7:30 - 9:00PM, Melbourne Aquarium
This Networking dinner will be held at the Melbourne Aquarium, King Street, Melbourne. If you wish to
attend please visit the registration desk for availability.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 20
Venue Map
Ground Floor
Level 1
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 21
Melbourne, Victoria
Melbourne is the Capital City of the State of Victoria. It has a population of approximately 3.5 million people.
Melbourne is commonly referred to as the cultural capital of Australia. It is the pre-eminent centre for arts and
culture, education, fine food and dining and shopping. Sport is also a city focus. Melbourne is home to many
of the world’s most famous sporting events F1 Grand Prix, Australian Tennis Open, AFL Grand Final and
Melbourne Cup Horse Race.
BANKING AND CREDIT CARDS
Most banks are open from 9.00am to 4.30pm from Monday to Thursday, banks are usually open until 5.00pm
on a Friday and some major banks are now open on a Saturday from 9.00am to 12.00pm. There are
Automatic Teller Machines (ATMs) located around the city which can be used after hours. Most banks are
able to cash travellers’ cheques.
Major credit cards are widely accepted. Your credit card can also be used at an ATM with your Personal
Identification Number.
CURRENCY EXCHANGE
There are many places to exchange currency in the CBD. Banks, travel agencies and dedicated exchange
spots are readily available.
TIPPING
Melbourne (or anywhere in Australia) does not operate on a tipping culture. You may tip to show your
appreciation for good service, but it is not a requirement.
TAXES IN MELBOURNE
A Goods and Services Tax of 10% tax is applied to most goods and services in Australia. All displayed prices
for Goods and Services are required to include the GST.
If you leave Australia within 30 days of buying goods, you may be exempt from paying GST. You can take
advantage of the Tourist Refund Scheme (TRS) to claim back the GST you have paid, and also claim back
any Wine Equalisation Tax (WET) you paid. TRS only applies to goods that you can carry as hand luggage,
and other conditions do apply.
CLIMATE
Spring is a mild period of year in Melbourne, the temperatures ranging from an average high of 20 degrees
Celsius to an average low of 10 degrees Celsius. September to November is the most changeable time of
the year for Melbourne as far as weather is concerned. The highest temperature on record has been beyond
25 degrees Celsius but there are lots of showers around and highs in temperature are rare.
TELEPHONE INFORMATION
If calling from a public telephone, the average price for a phone call to another landline within Australia is
50c. Phone cards can be purchased from post offices and newsagents, for local, interstate and international
calls. Mobile (cell phone) calls are more expensive.
When making international phone calls, the following formula should be used: 0011 + country code + area
code (if this has a ‘0’ please omit) + telephone number.
DINING OUT
Meals’ prices range anywhere from AU$10 at local cafes and small restaurants to AU$200 at world-leading
restaurants.
THINGS TO SEE AND DO
Melbourne has a multitude of inner city and regional tourist options that are easily accessible from the city.
From museums, plays, festivals and galleries to Federation Square, Crown Entertainment Complex and the
Eureka Skydeck. Other attractions include the Melbourne Aquarium, Great Ocean Road, and Queen Victoria
Markets. The following activities/tours are available for booking at the Registration Desk; Introduction to
Melbourne walk, Bike Ride, Yoga, Running, Golf.
TRANSPORT
Melbourne public transport system comprises of train, tram, ferries and bus services stretching across the
entire state of Victoria. The majority of Melbourne’s innercity experiences are walking distance from the
Convention Centre. Bike hire is also available throughout the inner city.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 22
Sponsor Listing
Merck Serono
Gold Sponsor
Website: www.merck.com.au
Merck Serono is the biopharmaceutical division of Merck. With headquarters in Darmstadt, Germany, Merck
Serono offers leading brands in 150 countries to help patients with cancer, multiple sclerosis, infertility,
endocrine and metabolic disorders as well as cardiovascular diseases. In the United States and Canada,
EMD Serono operates as a separately incorporated subsidiary of Merck Serono.
Merck Serono discovers, develops, manufactures and markets prescription medicines of both chemical and
biological origin in specialist indications. We have an enduring commitment to deliver novel therapies in our
core focus areas of neurology, oncology, immuno-oncology and immunology.
Nexvet
Gold Sponsor
Website: www.nexvet.com
Nexvet is a clinical-stage biopharmaceutical company focused on transforming therapeutics for companion
animals, such as dogs and cats, by developing and commercializing innovative biologics to address their key
unmet medical needs.
Nexvet’s proprietary PETization™ platform is designed to rapidly translate monoclonal antibodies between
species in a single step, enabling Nexvet to rapidly create “100% species-specific” therapeutics. Nexvet
believes this substantially reduces the time, cost and clinical risks associated with the conventional
development of biologics.
Through PETization, Nexvet has developed its lead biologic product candidates: the anti-nerve growth factor
(NGF) monoclonal antibodies (mAbs) NV-01 and NV-02 for the treatment of osteoarthritic pain in dogs and
osteoarthritic pain in cats, respectively. Further research has identified a tumor necrosis factor receptor
(TNFr) fusion protein, NV-08, for the treatment of chronic inflammatory diseases in dogs. Nexvet’s team
members have international track records in the development of human biologics and veterinary drugs, and
are committed to developing biologics to drive innovation in the global animal health therapeutic and vaccine
market, and give companion animals longer, happier lives.
Genentech
Silver Sponsor
Website: www.gene.com
Considered the founder of the industry, Genentech, now a member of the Roche Group, has been delivering
on the promise of biotechnology for over 35 years.
At Genentech, we use human genetic information to discover, develop, manufacture and commercialize
medicines to treat patients with serious or life-threatening medical conditions. Today, we are among the
world's leading biotech companies, with multiple products on the market and a promising development
pipeline.
MIMR-PHI Institute of Medical Research
Silver Sponsor
Website: www.mimr-phi.org
With a combined 75 years of research experience, more than 400 leading research experts and
postgraduate students and state-of-the-art research platforms and facilities, the Institute has taken its place
as a leader in improving global wellbeing through excellence in medical research and clinical application.
As one of Melbourne’s top medical research institutes and the research hub of the Monash Health
Translation Precinct (MHTP), MIMR-PHI scientists and clinicians are at the forefront of discovery and
translational research.
The Institute’s specialist Research Centres tackle key health priorities in the following areas: Cancer,
Genetic Disease, Immunity, Inflammation and Infectious Diseases, Reproductive Health, Fetal, Neonatal and
Children’s Health, Endocrinology and Metabolism.
Through our world-class research we strive to advance healthcare through an increased understanding of
disease and its diagnosis, treatment and prevention.
Pfizer’s Centers for Therapeutic Innovation
Silver Sponsor
Website: www.pfizer.com/research/rd_partnering/centers_for_therapeutic_innovation
The Centers for Therapeutic Innovation (CTI) is a pioneering research and development network initiated by
pharma that uses an open innovation model to bring great ideas to fruition. Part of Pfizer, we are an
entrepreneurial group that partners with leading academic medical centers and disease foundations with the
aim of translating promising science into clinical candidates.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 23
Our work is based on authentic collaboration, reflected in shared decision making and aligned incentives.
We are committed to bringing together cutting-edge academic and industry resources to develop medicines
faster and more efficiently.
With locations in Massachusetts, New York and California, our facilities are conveniently located on or near
academic campuses, allowing our partners to work closely with Pfizer scientists to translate research ideas
into clinical applications, with the ultimate goal of moving a therapeutic hypothesis through Proof-ofMechanism (PoM) in humans.
Through CTI, Pfizer is transforming the traditional model of drug discovery. By departing from the historical
pharma model of R&D, we are working to solve key challenges – namely the high cost and substantial time
investment – of drug discovery. CTI is rewiring the R&D ecosystem to offer new, targeted therapies to
patients living with disease.
Australian Biosearch / Bio Legend
Bronze Sponsor
Website: www.aust-biosearch.com.au / www.biolegend.com
BioLegend, with their Australian partner, Australian Biosearch, provide world-class antibodies, proteins,
assays, and custom solutions. Cytokine products include a Multiplex solution with new LEGENDplex™ MultiAnalyte Flow Assay Kits, Recombinant Cytokines/Chemokines and LEAF™ antibodies for in vivo assays
(Bulk pricing available). New LEGEND MAX™ ELISA Kits: TGF-β family (total, free active, latent, LAP,
Soluble GARP); CCL8, Adiponectin, Free SCF, FGF-basic. New PE/Dazzle™ 594 conjugates for Multicolor
Flo
LEGENDScreen™ Cell Screening Kits and Zombie dyes for live/dead cell discrimination. With our new
Neuroscience R&D team and products, BioLegend is positioned to become a leader in neuroscience and
immunology research.
Bio-Rad Laboratories
Bronze Sponsor
Website: www.bio-rad.com
Bio-Rad Laboratories has been at the centre of scientific discovery for 60 years, developing products and
solutions to advance healthcare and research.
Multiplex ELISA on magnetic bead xMAP technology enables the detection and measurement of up to 100
analytes in a single sample. Cytokines have great utility as biomarkers for disease including cancer and
autoimmunity; therefore multiplexing gives you the power to understand complex relationships of proteins in
immunological pathways. Bio-Rad’s Bio-Plex assays are designed to quantitate proteins in diverse matrices
such as serum, plasma and supernatant at small sample volumes. Disease state biomarker panels and
pathways available include: Cancer, Apoptosis and Metabolism.
Visit the Bio-Rad booth to learn how this technology could be applied to your research.
Bio-Techne
Bronze Sponsor
Website: www.bio-techne.com
Bio-Techne brings together the world class brands of R&D Systems, Novus Biologicals, and Tocris to better
serve our customers and the community. R&D Systems cytokines defined the industry 30 years ago and
continue to provide researchers with reliable research results. As the Bio-Techne portfolio grows, we are no
longer just about consumables. We are adding innovative instruments to our portfolio in a way that allows us
to leverage our gold standard reagents into complete solutions for both life science and diagnostic markets.
Visit us at booth #8 to learn more about the entire Bio-Techne product portfolio.
eBioscience
Bronze Sponsor
Website: www.ebioscience.com
eBioscience, an Affymetrix company, develops and manufactures over 11,000 antibodies, proteins,
immunoassays and multiplex assays at ISO-certified facilities worldwide. Focused on accelerating scientific
discovery in immunology and oncology, we provide innovative solutions to researchers and clinicians looking
to answer questions driving today's life science communities. Partner with the industry leader of translational
science.
PBL Assay Science
Bronze Sponsor
Website: www.pblassaysci.com
PBL Assay Science, your trusted source for interferon ELISAs, proteins, antibodies, and assay services has
expanded its human cell-expressed cytokine and growth factor line and ultrasensitive cytokine detection
services. We invite you to visit with our scientists at Booth 7 in the ICIS exhibition space to find out how we
can help you address your most pressing assay challenges. Whether you would like us to customize a
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 24
multiplex assay incorporating your key analytes, to serve as an outside laboratory to run samples in
confirmatory assays, or to provide a novel human cell-expressed protein, PBL is here to serve your needs.
Elsevier
Sponsor
Website: www.elsevier.com
Elsevier is a world-leading provider of information solutions that enhance the performance of science, health,
and technology professionals. We publish nearly 2,200 journals, including a prestigious collection of
immunology journals, The Lancet and Cell, and more than 25,000 book titles. Visit elsevier.com for more
information.
Opsona Therapeutics
Session Sponsor
Website: www.opsona.com
Opsona Therapeutics is a leading immunology drug development company, focused on novel therapeutic
approaches to key targets of the innate immune system associated with a wide range of major human
diseases, including autoimmune and inflammatory diseases, with specific focus on Solid Organ
Transplantation and Oncology.
The company was founded in 2004 by three world-renowned immunologists at Trinity College in Dublin.
Opsona's lead product, a fully humanized monoclonal IgG4 antibody (OPN-305) targeting Toll-like-receptor-2
(TLR2) has demonstrated activity in a number of preclinical models and has been tested in a Phase I clinical
trial in healthy volunteers. The company has initiated a three-part multi-centred, double blinded and placebo
controlled Phase II clinical study to evaluate the safety, tolerability and efficacy of OPN-305 in renal
transplant patients at high risk of Delayed Graft Function (DGF) as the first clinical indication for the
development of OPN-305 of which the first part has been completed successfully. OPN-305 has obtained
EMA and FDA orphan drug status in Solid Organ Transplantation.
Further information is available at: http://www.opsona.com/.
Mary Ann Liebert Inc.
Sponsor
Website: www.liebertpub.com/jicr
Journal of Interferon and Cytokine Research (JICR), led by Co-Editors-in-Chief Ganes C. Sen and Thomas
A. Hamilton, celebrates its 35th anniversary in 2015. Over the last three and a half decades the Journal has
been a major source of new primary articles covering IFN-mediated anti-viral mechanisms and topical
reviews covering important emerging areas by top contributors in the field of IFN and cytokine research. The
relevance of such articles is reflected in our impact factor of 3.899, representing an 18% increase over last
year and the 5th consecutive year of growth.
Walter & Eliza Hall Institute of Medical Research
Sponsor
Website: www.wehi.edu.au
The Walter and Eliza Hall Institute is Australia’s oldest medical research institute and will celebrate its
centenary in 2015. Affiliated with The University of Melbourne and The Royal Melbourne Hospital, the
institute is home to more than 750 researchers working on cancers, infectious diseases and immune
disorders.
The institute has more than 100 scientists working on cytokine signalling. The institute’s scientists were
instrumental in the discovery of many cytokines, cytokine receptors and the suppressors of cytokine
signalling (SOCS) proteins, and their functions. Current projects expand on this work, investigating the role
of cytokines and their suppressors in many diseases including blood cancers, gastrointestinal cancer,
rheumatoid arthritis and influenza.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 25
Exhibitor Map & Listing
Abcam Australia
Booth 7
Website: www.abcam.com
Abcam is a provider of high quality protein research tools. We are delighted to launch the direct service in
Australia and New Zealand since August 2014. Researchers can benefit from:
- Direct technical support
- Improved delivery – reduced by up to 50% from order to delivery
- Special local events and webinars
Australian Biosearch / BioLegend
See Sponsorship Listing for details.
Booth 9
Bio-Rad Laboratories
See Sponsorship Listing for details.
Booth 6
Bio-Techne
See Sponsorship Listing for details.
Booth 8
eBioscience
See Sponsorship Listing for details.
Booth 4
Lonza Australia
Booth 2
Website: www.lonza.com
Lonza is a leading supplier of human normal and diseased cells, cell culture media, cell based assays and
transfection devices and reagents and Endotoxin Detection devices, reagents and services to the
pharmaceutical, healthcare and life science industries from research to final product manufacture.
Provider of Endotoxin Detection Assays, Rapid Microbial Detection Technology, Nucleofector™ Technology,
Clonetics™ and Poietics™ Cells and Media, BioWhittaker™ Media, FlashGel™ System, PAGEr™ Precast
Gels, and SeaKem® Agarose.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 26
Miltenyi Biotec Australia
Booth 3
Website: www.miltenyibiotec.com
Miltenyi Biotec & MACS® technology is the global market leader in magnetic cell separation. We offer
complete workflow solutions from sample preparation to cell separation, media and cytokines for cell culture,
flow cytometry and small animal pre-clinical imaging. Miltenyi Biotec provides expert technical advice in a
wide range of scientific fields including, amongst others: immunology, cancer, stem cells and neuroscience.
We offer GMP cellular therapeutic products and are committed to the promise of cellular medicine in the
fields of organ regeneration, immune modulation and transplantation.
Phone: +61 (2) 8877 7400
Email: [email protected]
PBL Assay Science
See Sponsorship Listing for details.
Booth 5
Peprotech
Booth 1
Website: www.peprotech.com
Since 1988 PeproTech has been producing a wide range of recombinant proteins, mainly Cytokines, their
related Antibodies and ELISA Kits; all of the highest quality and at the most competitive price. PeproTech
appreciates the opportunity to support the ISCT 2014, and is keen to learn more about the scientific
community and strengthen their level of cooperation.
Please visit the PeroTech Stand to receive your Cell Reprogramming related Factors comprehensive
booklet, Information on ESC/iPSC characterization kit, the PeproTech ELISA Development kits and much,
much more. Or just come to the stand for a chat and to play a game to WIN!
The PeproTech Australia office is managed by LONZA Australia, Ph.1300 657 508 or contact: Margret
Schuller; 0430353301; [email protected]
Transnetyx
Booth 10
Website: www.transnetyx.com
Transnetyx Automated Genotyping is fast, easy, and accurate and believes in making things simple. With
more than 50 million successful reactions globally, sending your genotyping to Transnetyx is designed to
eliminate the tedious process of extracting and testing DNA so researchers and labs can get back to what is
most important - their research. Eliminate overcrowded cages and high cage costs with the efficiency and
reliability of Transnetyx. Genotyping has evolved. Have you?
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 27
Program – Sunday 26th October 2014
11:00-17:00
ICIS Council and Committee Meetings
14:00-20:00
REGISTRATION
17:30-19:30
OPENING SESSION
Rooms 101 and 102
Level 1 Foyer / Main Foyer
Plenary 1
17:30-17:55
Opening Remarks (Brendan Jenkins, Co-convenor)
Award Presentations (Richard Flavell, President, ICIS)
17:55-18:20
ICIS Honorary Lifetime Membership Awards
Chair: Richard Flavell
Amanda Proudfoot, NovImmune, Switzerland
“Targeting chemokines: Pathogens can, why can’t we?” abs# 318
Chuck Samuel (presentation to be given in Concurrent Symposia 6)
18:20-18:45
Seymour and Vivian Milstein Award for Excellence in Interferon and
Cytokine Research
Chair: Richard Flavell
Kate Fitzgerald, University of Massachusetts Medical School, USA
“Regulation of the Interferon Response by the long non-coding RNA lincRNAEPS” abs# 249
18:45-19:30
Keynote lecture 1
Chair: Paul Hertzog
Nicos Nicola, Walter and Eliza Hall Institute, Australia
“Old and new families of negative regulators of cytokine signalling” abs# S-37
19:30-21:00
WELCOME RECEPTION
Level 1 Foyer / Main Foyer
Including indigenous dance performance
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 28
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 29
Program – Monday 27th October 2014
08:00-18:00
REGISTRATION
08:30-10:30
PLENARY SESSION 1
Level 1 Foyer / Main Foyer
Plenary 1
Cytokines in the control of innate and adaptive immunity
(Sponsored by Walter & Eliza Hall Institute of Medical Research, Australia)
Chairs: Matt Sweet, Dhan Kalvakolanu
08:30-09:00
Zhijian James Chen, University of Texas Southwestern Medical Center, USA
“Innate immune sensing and signaling of cytosolic DNA and RNA” abs# S-8
09:00-09:30
Fabienne MacKay, Monash University, Australia
“Cytokine-driven loss of plasmacytoid dendritic cell function in chronic
lymphocytic leukemia” abs# S-15
09:30-10:00
Zhengfan Jiang, Peking University, China
“Identification and Characterization of Phosphodiesterase V-cGAPs That
Degrade 3’3’-Cyclic GMP-AMP in vibrio cholerae" abs# S-33
10:00-10:15
Julia Ellyard, Australia National University, Australia
“Cytokine dysregulation in autoimmune disease pathogenesis” abs# 246
10:15-10:30
Hong Tang, Chinese Academy of Sciences, China
“Innate cell-like function of T cells in modulation of inflammatory response to
infection and tissue injury” abs# 335
10:30-11:00
MORNING TEA BREAK
11:00-13:00
CONCURRENT SYMPOSIA 1
Level 1 Foyer / Main Foyer
Rooms 105/106
Cytokines and the regulation of tissue resident immune cells
(Sponsored by PBL Assay Science, Australia)
Chairs: Iain Campbell, Catarina Sacristan
11:00-11:30
Frederic Geissman, King’s College, UK
“Origin, development and maintenance of tissue resident macrophages” abs# S-
30
11:30-12:00
Florent Ginhoux, Singapore Immunology Network, Singapore
“Macrophage and dendritic cell ontogenuy” abs# S-31
12:00-12:15
Dominic De Nardo, Institute of Innate Immunity, Germany (ICIS Young
Investigator Awardee)
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 30
“Modulation of innate immune responses by ATF3” abs# 36
12:15-12:30
Margaret Hibbs, Monash University, Australia
“G-CSF is a critical mediator of chronic inflammatory lung disease” abs# 264
12:30-12:45
Cathleen Pfefferkorn, University Medical Center Freiburg, Germany
“Identification of interferon-ß-producing cells in the virus-infected brain” abs# 148
12:45-13:00
Ian Humphreys, Cardiff University, UK
“Neutrophils recruited by IL-22 in peripheral tissues function as TRAIL-dependent
antiviral effectors against MCMV” abs# 87
11:00-13:00
CONCURRENT SYMPOSIA 2
Plenary 1
The microbiome and mucosal immune responses
(Sponsored by e-Bioscience, an Affymetrix Company, Australia)
Chairs: Richard Ferrero, Thirumula-Devi Kanneganti
11:00-11:30
Richard Flavell, Yale University School of Medicine, USA
“Inflammasomes in health, dysbiosis, and disease” abs# S-11
11:30-12:00
Gabriel Nunez, University of Michigan, USA
"Linking pathogen virulence, immunity and the microbiota" abs# S-18
12:00-12:30
Elizabeth Hartland, University of Melbourne, Australia
“Death receptors and bacterial diarrhoea” abs# S-12
12:30-12:45
Phil Hansbro, University of Newcastle, Australia
“Shifts in the microbiome during experimental chronic obstructive pulmonary
disease (COPD)” abs# 261
12:45-13:00
Tomonori Kamiya, Tokyo University of Science, Japan
“Dectin-1
modifies
colonic
microflora
by
inducing
cytokine-dependent
antimicrobial peptide secretion from intestinal epithelial cells” abs# 275
13:00-14:00
LUNCH BREAK (lunch provided)
Level 1 Foyer / Main Foyer
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 31
14:00-15:30
CONCURRENT MINI-SYMPOSIUM 1
Plenary 1
Chairs: Claudia Nold, Rose Ffrench
14:00-14:15
Stacy Horner, Duke University Medical Center, USA (ICIS Young Investigator
Awardee)
“Proteomic analysis of mitochondrial-associated ER membranes during RNA
virus infection reveals dynamic changes in protein and organelle trafficking” abs#
267
14:15-14:30
Florian Wiede, Monash University, Australia
“The role of PTPN2 in early thymocyte development and JAK/STAT signaling”
14:30-14:45
Andrew Larner, Virginia Commonwealth University, USA abs# 351
“STAT3 interacts with Cyclophilin D in cancer cells to regulate the mitochondrial
permeability transition pore” abs# 107
14:45-15:00
Saleela Ruwanpura, MIMR-PHI Institute, Australia
“Novel role for interleukin-6 trans-signaling in the pathogenesis of pulmonary
inflammation and emphysema” abs# 161
15:00-15:15
Rebecca Coll, University of Queensland, Australia
“MCC950 is a potent and specific inhibitor of the NLRP3 inflammasome and a
novel potential therapeutic for NLRP3 driven diseases” abs# 30
15:15-15:30
Amanda Huber, University of Michigan, USA
(Sidney & Joan Pestka Post-Graduate Award in Interferon & Cytokine
Research Sponsored by PBL Interferon Source)
“The Type I Interferon axis suppresses Th-17 driven autoimmune inflammation in
a mouse model of Multiple Sclerosis via Interferon Regulatory Factor-7” abs# 86
14:00-15:30
CONCURRENT MINI-SYMPOSIUM 2
Rooms 105/106
Chairs: Niamh Mangam, Zoltan Fehervari
14:00-14:15
Yeonseok Chung, Seoul National University, South Korea (Journal of Biological
Chemistry/Herbert Tabor Young Investigator Award) abs# 233
“Proatherogenic Condition Promotes Autoimmune Th17 Cell Responses”
14:15-14:30
Laura Dagley, Walter and Eliza Hall Institute, Australia
“A mass spectrometry-based approach for studying kinase activity” abs# 33
14:30-14:45
Sonja Best, NIAID/NIH, USA
“Viral antagonism of type I interferon responses reveals prolidase as a regulator
of IFNAR1 trafficking and expression” abs# 220
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 32
14:45-15:00
Sukanya Raghaven, University of Gothenburg, Sweden
“The role of IL-17A and IFNγ in vaccine-induced protection against Helicobacter
pylori infection” abs# 154
15:00-15:15
Betsy Barnes, Rutgers Biomedical and Health Sciences, USA
“Intrinsic and extrinsic mechanisms of metastatic inhibition by IRF5 in human
ductal carcinoma” abs# 218
15:15-15:30
Michael White, Walter and Eliza Hall Institute, Australia
“The apoptotic caspase cascade suppresses mitochondrial DNA-induced STINGmediated type I IFN production by dying cells” abs# 197
15:30-16:00
AFTERNOON TEA BREAK
16:00-18:00
CONCURRENT SYMPOSIA 3
Inflammasomes and the IL-1 cytokine family
Level 1 Foyer / Main Foyer
Plenary 1
(Sponsored by BioLegend/Australian Bioresearch, Australia)
Chairs: Ashley Mansell, Betsy Barnes
16:00-16:30
Thirumala-Devi Kanneganti, St Jude Children's Research Hospital, USA
“IL-1 regulation in inflammatory disease” abs# S-34
16:30-17:00
Denise Monack, Stanford University, USA
“Host recognition of intracellular bacterial pathogens” abs# S-17
17:00-17:30
Veit Hornung, University of Bonn, Germany
“Cytosolic recognition of nucleic acids by the innate immune system” abs# S-32
17:30-17:45
Seth Masters, Walter and Eliza Hall Institute, Australia (ICIS Young Investigator
Awardee)
“Inflammasome activation due to polymerized actin triggers an autoinflammatory
disease that is dependent on IL-18, not IL-1β” abs# 126
17:45-18:00
Kate Schroder, University of Queensland, Australia (ICIS Young Investigator
Awardee)
“The neutrophil NLRC4 inflammasome selectively promotes IL-1β maturation
without pyroptosis during acute Salmonella challenge” abs# 165
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 33
16:00-18:00
CONCURRENT SYMPOSIA 4
Rooms 105/106
Cytokine signalling mechanisms
(Sponsored by Nexvet Biopharma, Australia)
Chairs: Sandra Nicholson, George Stark
16:00-16:30
Uwe Vinkemeier, University of Nottingham, UK
“STAT protein phase transitions in cytokine signalling” abs# S-26
16:30-17:00
Jeff Babon, Walter and Eliza Hall Institute, Australia
“Inhibition of IL-6 family cytokines by SOCS3: characterizing the mode of
inhibition and the basis of its specificity” abs# S-3
17:00-17:30
Andrew Brooks, University of Queensland, Australia
“A new cytokine receptor activation paradigm: activation of JAK2 by the growth
hormone receptor” abs# S-7
17:30-17:45
Sophie Broughton, St Vincent’s Institute, Australia (Christina Fleischmann
Award to Young Women Investigators)
“Structural and functional analysis of the GM-CSF:GM-CSF receptor alpha chain
binary complex provide new insights into signalling of the GM-CSF ternary
complex” abs# 17
17:45-18:00
Marcel Nold, MIMR-PHI Institute, Australia
“IL-37 requires IL-18Rα and SIGIRR to carry out its multi-faceted antiinflammatory program on innate signal transduction” abs# 308
18:00-19:30
POSTER SESSION 1
Level 1 Foyer / Main Foyer
Including trade/exhibitor display with drinks and cheese
19:30-21:00
Networking mixer for students and ECR’s
The Boatbuilders Yard
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 34
Program – Tuesday 28th October 2014
08:00-18:00
REGISTRATION
08:30-10:30
PLENARY SESSION 2
Level 1 Foyer / Main Foyer
Plenary 1
Cytokines and interferons in cancer
(Sponsored by Merck Group, Germany)
Chairs: Brendan Jenkins, Tracy Putoczki
08:30-09:00
Frances Balkwill, Barts Cancer Institute, UK
“Targeting inflammatory cytokines and chemokines in cancer” abs# S-4
09:00-09:30
Mark Smyth, Queensland Institute of Medical Research, Australia
“Checkpoints and interferons in tumor control” abs# S-24
09:30-10:00
Belinda Parker, La Trobe University, Australia
”Exploiting the type-1 interferon pathway as a biomarker and therapeutic target
for metastatic cancer” abs# S-20
10:00-10:30
Masaaki Murakami, Osaka University, Japan
“The inflammation amplification loop in non-immune cells, which is regulated by
cytokines and neural activation” abs# S-36
10:30-11:00
MORNING TEA BREAK
11:00-13:00
CONCURRENT SYMPOSIA 5
Level 1 Foyer / Main Foyer
Rooms 105/106
Innate lymphoid cells and cytokine secretory mechanisms
(Sponsored by Bio-Rad Laboratories, Australia)
Chairs: Meredith O’Keefe, Howard Young
11:00-11:30
Dale Umetsu, Genentech, USA
“A role for ILC2s, ILC3s and the NLRP3 inflammasome in different forms of
asthma” abs# S-25
11:30-12:00
Gabrielle Belz, Walter and Eliza Hall Institute, Australia
“Critical mucosal protection by the complex innate lymphoid cell network” abs# S-
5
12:00-12:30
Wolfgang Weninger, Centenary Institute, Australia
“Regulation of inflammation by perivascular macrophages” abs# S-40
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 35
12:30-12:45
Elena Vigano, Singapore Immunology Network, Singapore
“A novel player for the regulation of IL-1α/β release in human monocytes” abs#
189
12:45-13:00
Gregory Bouchaud, French National Institute for Agricultural Research, France
“Previous food allergy aggravates allergic markers and intestinal damages in a
mouse model of asthma” abs# 14
11:00-13:00
CONCURRENT SYMPOSIA 6
Plenary 1
Mary Ann Liebert, Inc. Symposium in Honour of Philip I. Marcus
Chairs: David Levy, Paula Pitha-Rowe
11:00-11:45
Chuck Samuel, University of California Santa Barbara, USA
ICIS Honorary Lifetime Membership Awardee
“RNA adenosine deaminase ADAR1 and RNA-dependent protein kinase PKR:
opposing modulators of antiviral innate immunity” abs# S-22
11:45-12:15
Peter Staehli, Institute for Virology, Germany
“Epithelial IFN-λ and IFN-α/β constitute a compartmentalized mucosal defense
system that restricts virus infection of the intestinal and respiratory tract” abs#
333
12:15-12:30
Annie Bruns, Northwestern University, USA
(Sidney & Joan Pestka Graduate Award in Interferon & Cytokine Research
Sponsored by PBL Interferon Source)
“The innate immune sensor LGP2 activates antiviral signaling by regulating
MDA5-RNA interaction and filament assembly” abs# 229
12:30-12:45
Julia Heinrich, Institute for Experimental Infection Research, Germany
“Kinetics of Type I Interferon responses determine either establishment of
antiviral memory or no induction of adaptive immunity” abs# 77
12:45-13:00
Nathalie Grandvaux, University of Montreal, Canada
“Identification of two distinct mechanisms that control the duration of the
Interferon-mediated antiviral response” abs# 64
13:00-16:00
LUNCH BREAK (lunch provided; also Melbourne City Tours available)
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 36
16:00-18:00
CONCURRENT SYMPOSIA 7
Plenary 1
Genomic regulation of cytokine and interferon responses
Chairs: Chuck Samuel, Kate Fitzgerald
16:00-16:30
K Mark Ansel, University of California San Francisco, USA
“MicroRNA regulation of T cell cytokine responses” abs# S-2
16:30-17:00
Xia-Fang Wang, Duke University, USA
“Tumor microenvironment: microRNAs and metastasis” abs# S-39
17:00-17:15
Colleen Elso, St Vincent’s Institute, Australia
“Apics deficiency reveals a role for a long noncoding RNA in dendritic cell
function and autoimmunity” abs# 42
17:15-17:30
Isabelle Marie, NYU School of Medicine, USA
“Positive and Negative Epigenetic Regulatory Steps During IFN-Stimulated
Transcriptional Initiation and Elongation” abs# 297
17:30-17:45
Michael Gantier, MIMR-PHI Institute, Australia
“Master regulation of transcriptional responses by microRNAs” abs# 253
17:45-18:00
Kasia Blaszczyk, Mickiewicz University, Poland (Melbourne Travel Awardee)
“IFNɑ engages a sustained antiviral response depending on STAT2/IRF9 but not
ISGF3” abs# 223
16:00-18:00
CONCURRENT SYMPOSIA 8
Rooms 105/106
Metabolic homeostasis and autophagy
Chairs: Bryan Williams, Carolyn Geczy
16:00-16:30
Vojo Deretic, University of New Mexico, USA
”Immunological manifestations of autophagy” abs# S-29
16:30-17:00
Mark Febbraio, Baker Institute, Australia
“Fatty acid-induced inflammation in immune cells: implications for metabolic
disease” abs# S-10
17:00-17:15
Maria
Kaparakis-Liaskos,
MIMR-PHI
Institute,
Australia
(ICIS
Young
Investigator Awardee)
“Identification of the intracellular location and mechanisms of NOD1-dependent
inflammatory responses” abs# 93
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 37
17:15-17:30
Eleanor Fish, University of Toronto, Canada
“IFN-ß regulation of glucose metabolism is PI3K/Akt dependent and important for
antiviral activity against coxsackievirus B3” abs# 55
17:30-17:45
Dhan Kalvakolanu, University of Maryland Baltimore, USA
“Interferon-induced autophagy and antibacterial defenses occur through the
Apoptosis stimulating kinase-1” abs# 273
17:45-18:00
James Harris, Monash University, Australia
“Autophagic Regulation of Pro-inflammatory Cytokines” abs# 262
18:00-19:30
POSTER SESSION 2
Level 1 Foyer / Main Foyer
Including trade/exhibitor display with drinks and cheese
19:30-21:30
NETWORKING FUNCTION
Melbourne Aquarium
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 38
Program – Wednesday 29th October 2014
08:00-17:30
REGISTRATION
08:30-10:30
PLENARY SESSION 3
Level 1 Foyer / Main Foyer
Plenary 1
Cytokines and interferons in the inflammatory response
(Sponsored by MIMR-PHI Institute of Medical Research, Australia)
Chairs: John Hamilton, Kate Stacy
08:30-09:00
Yinon Ben-Neriah, The Hebrew University of Jerusalem, Israel
“Parainflammation in cancer“abs# S-6
09:00-09:30
Elina I Zuniga, University of California San Diego, USA
“Dynamics of cytokine responses during chronic viral infection” abs# S-35
09:30-10:00
Simon Jones, Cardiff University, UK
“Interleukin-6 drives tissue fibrosis in response to repeat acute inflammation”
abs# S-13
10:00-10:30
Kingston Mills, Trinity College, Ireland
“Function and regulation of IL-17 cytokine family in infection and autoimmunity”
abs# S-16
10:30-11:00
MORNING TEA BREAK
Level 1 Foyer / Main Foyer
11:00-12:00
CONCURRENT MINI-SYMPOSIUM 3
Plenary 1
Chairs: Lisa Mielke, Robert Silverman
11:00-11:15
Helene Minyi Liu, National Taiwan University, Taiwan (Melbourne Travel
Awardee)
“Deacetylation-dependent
regulation
of
RIG-I
activation
by
the
HDAc6
deacetylase mediates innate anti-viral immunity” abs# 113
11:15-11:30
Cristina Bergamaschi, National Cancer Institute at Frederick, USA
“Heterodimeric IL-15 promotes tumor control through the regulation of the
balance of effector and regulatory cells via an IL-2 deprivation mechanism” abs#
8
11:30-11:45
Tim Hercus, Centre for Cancer Biology, Australia
“Insights into the function of an anti-leukaemia antibody: structural studies of
CSL362 bound to soluble CD123” abs# 78
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 39
11:45-12:00
Tao Lu, Indiana University, USA
“PRMT5 dimethylates R30 of the p65 subunit to activate NF-κB” abs# 120
11:00-12:00
CONCURRENT MINI-SYMPOSIUM 4
Rooms 105/106
Chairs: Patricia Fitzgerald-Bocarsly, Benjamin Kile
11:00-11:15
Matthias Parrini, University of Veterinary Medicine, Vienna, Austria (Melbourne
Travel Awardee)
“STAT1 Isoforms in Transcriptional Control – Distinct Traits” abs# 144
11:15-11:30
Daniel Gough, MIMR-PHI Institute, Australia
“STAT3 supports experimental K-RasG12D-induced murine myeloproliferative
neoplasms dependent on serine phosphorylation” abs# 63
11:30-11:45
Nancy Reich, Stony Brook University, USA
“Gammaherpesvirus requires STAT3 to establish B cell viral latency in animals”
abs# 158
11:45-12:00
Howard Young, National Cancer Institute, USA
“Inhibiting cytokine activity – the use of modified DNA aptamers as an alternative
to neutralizing antibodies” abs# 208
12:00-13:00
LUNCH BREAK (lunch provided)
13:00-15:00
CONCURRENT SYMPOSIA 9
Plenary 1
Pattern recognition receptors and responses to pathogens
(Sponsored by bio-Techne, Australia)
Chairs: Scott Durum, Eleanor Fish
13:00-13:30
Shizuo Akira, Osaka University, Japan
“Regnase-1, a ribonuclease involved in the inflammatory and immune responses”
abs# S-1
13:30-14:00
Eicke Latz, University of Bonn, Germany
“Inflammasomes have extracellular activities that propagate inflammation” abs#
S-14
14:00-14:30
Feng Shao, National Institute of Biological Sciences, China
“Innate immune sensing of bacteria and bacterial virulence by the cytosolic
inflammasome complex” abs# S-23
14:30-14:45
Adam Wall, University of Queensland, Australia
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 40
“Regulation of inflammatory cytokine responses by a new TLR4 regulator
complex of Rab8a/PI3Kγ” abs# 347
14:45-15:00
Ashley Mansell, MIMR-PHI Institute, Australia
“PB1-F2 from H7N9 Influenza A virus activates the NLRP3 inflammasome to
induce inflammation” abs# 296
13:00-15:00
CONCURRENT SYMPOSIA 10
Rooms 105/106
Translatable outcomes targeting cytokines: the lessons learned
(Sponsored by Nexvet Biopharma, Australia)
Chairs: Amanda Proudfoot, Simon Jones
13:00-13:30
Daniel Cua, Merck Research Laboratories, USA
“IL-23 regulation of innate and adaptive immunity” abs# S-28
13:30-14:00
Stefan Rose-John, University of Kiel, Germany
“Blocking only the bad side of Interleukin-6” abs# S-21
14:00-14:30
Hua Yu, Beckman Research Institute, USA
“STAT3: from fundamental discoveries to the clinic” abs# S-27
14:30-14:45
Walter Ferlin, NovImmune, Switzerland
“Exploiting receptor clustering enhances inhibitory effects of a therapeutic
monoclonal antibody to TLR4 revealing a novel mechanism of action driving the
pathology of rheumatoid arthritis” abs# 51
14:45-15:00
Scott Durum, National Cancer Institute, USA
“IL-7R: a target in ALL and autoimmunity” abs# 245
15:00-15:30
AFTERNOON TEA BREAK
Level 1 Foyer / Main Foyer
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 41
15:30-17:15
CLOSING SESSION
(Sponsored by Opsona Therapeutics, Ireland)
15:30-16:15
Keynote lecture 2
Chair: Elizabeth Hartland
Peter Doherty, University of Melbourne, Australia
“Cytokine profiles in experimental and human influenza” abs# S-9
16:15-17:00
Keynote lecture 3
Chair: Paul Hertzog
Luke O’Neill, Trinity College, Ireland
“A lifelong love affair with IL-1 and inflammation” abs# S-38
17:00-17:15
Closing remarks (Paul Hertzog, Co-convenor)
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 42
Plenary 1
Abstracts
Additional Abstracts:
Please refer to “Cytokine”, provided in your satchel, for speaker abstracts S-1 – S-27 and meeting abstracts
1 – 212. Abstracts are numbered in alphabetical order by the presenting author surname.
Withdrawn:
Abs#1
Abs#40
Abs#56
Abs#57
Abs#58
Abs#61
Abs#111
Abs#114
Abs#134
Abs#137
Abs#155
Abs#183
Abs#192
Abs#215
Author Index
Abankwa, D
S-7
Abbott, C
303
Abdirahman, S
A Andrews, A
164,20,254
0
Angel, C
279
Barry, E.F
276,78
213
Angelovich, T.A
215
Baschuk, N
321,S-20
Abdul Hay, M
287
Ansel, K
S-2
Bathke, B
266
Abe, R
339
Antes, A
340
Baturcam, E
117,6
Abramovich, R
73
Arakelova, E
226
Beach, R.K
8
Achard, M.E
277
Aran, D
S-6
Bear, J
8
Aderem, A
152
Ariffin, J.K
216
Becher, D
194
Afonina, I
2
Arneborg, N
170
Bedard, K.M
118
Agarwal, A
349
Arumugam, T.V
119
Beddoe, T
242
Aharoni, R
73
Ashton, M.P
42
Bedoui, S
197,7
Ahmad, F
97
Aubert, P
11,14
Beilharz, M.W
101
Ahmed, A.U
3
Aubert, V
75
Beker, F
285
Ahn, M
269
Aukrust, P
314
Belgnaoui, S
116
Ahn, Y
102
Aw, J
329
Bell, T
133
Aifantis, I
287,63
Ayllon, J
326
228
Akira, S
S-1
Azam, T
308
Alekseev, A
310
Bell, T.D
Bellows-Peterson,
M.L
Alexander, K.A
188
Alexander, W
281,286,30
1,S-37
Ali Naqvi, R
Bae, Y
33,307,S-3,
S-37
272
Baglaenko, Y
217
180
Baker, A
170
Alicea, C
8
101
Allam, R
301
Allegood, J.C
283
Bakker, E
Balasubramanian,
S
Balinsky, C.A
Allen, A
S-16
Almo, S.C
340
Amalraj, J
202,322,
345
320
Andersen, L
252
Almolda, B
Anderson, R.L
139,161,26
4
321
Anderton, H
286,47
Anderson, G.P
Babon, J
260
Benedict, C.A
130,278,30
6,313,S-5
87
Ben-Neriah, Y
S-6
Bergamaschi, C
8
Berger, P
309
Berger, P.J
231
4
Berges, J
48
Balkwill, F
S-4
Bernard, C
42
Bandeira, A
48
Bernardino, P.N
31,162
Bär, E
350
Bertheloot, D
9
Baran, M
5
Bertin, J
93
Barata, J.T
245
Best, A
221
Bardin, P.G
184,337
Best, S.M
220
Barletta, R.G
323
Beyaert, R
2
Barnes, B
218,219,
240,241,25
Bhardwaj, R
283
Bhaskar, K
10
344
Belz, G
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 43
Bhattacharyya, A
222
Brooks, G.D
16
Chabauty, J
11
Bibert, S
BielefeldtOhmann, H
Bihouee, T
75
Broughton, B.R
119
Chae, J
126,30
Broughton, S.E
17,276,78
Chae, J.J
237
Brown, E.J
169
Chakrabarti, A
205
278
Brown, E.L
200
Chakravorty, A
302
Bird, N
340
Brown, J
243
Chan, K.L
23
Birge, R.B
Brown, L
Chang, H
297
Bishai, W.
135
296
222
Browning, A.F
18
Chang, M.W
24
Biswas, S
223
Bruns, A.M
229
Chang, N
217
Blaszczyk, K
188
Buatois, V
49,50
Chang, T
25
Blazar, B.R
Buchert, M
Chang, Y
S-25
Blondel, D
133,228
150
S-7
Budin, S
89
Chaplin, P
266
Blucher, K.M
223
Bufler, P
308
Chapman, R
230
Bluijssen, H
75
Bulani, V
100,19
Chatel, L
49
Bochud, P
Bunting, M
Chatfield, S
254
Bodinier, M
11,14
295
220
Burger, D
227
Chatzinikolaou, A
200
Boehm, M
136
Burke, D
55
Cheminant, M
14
Bogoyevitch, M.A
277
Burns, C
33,305
Chen, K
165
Bokil, N.J
Burugu, S
Chen, S
107,8
Bonanzinga, S
224
34
93
Busfield, S
78,164,254
Chen, X
201,255
Boneca, I
248
Busuttil, R
306
Chen, Y
223,254,26
Boog, B
12
Buzalaf, M.A
129
Chen, Z
S-8
Borst, K
Calarese, D.A
Cheng, W
231
Boshuizen, M.C
13
340
11,14
Cambier, J
197
Cheong, M
188
Bouchaud, G
225
Caminschi, I
191
Chertova, E
8
Bouillet, P
133
Chew, C
125
Chhabra, Y
S-7
Bourke, N
306
Chiang, J
307
Boussioutas, A
5
Chin, K
168
Bowie, A
Ching, A
S-37
Boyajyan, A
226
Cao, X
20,84,91,
202,255,
257,322,
257,332,
334,345
67
27
Bourhy, H
52
Chiramel, A.I
220
Boyd, R
351
Carbone, F.R
35
295,320
Cardenas, K
Chmielewski, S
223
boyle, g
Cho, M
142
Bozinovski, S
139
Cardoso, B.A
245
Cho, S
309
Branchett, W
260
Cardoso, E.O
31,162
186
Cho, S.X
231,308
Brandalise, S.R
245
Cardus, A
349
227
Carey, A.B
Choi, G
28
Brandt, K.J
Choi, I
102,271
Braniff, S
242
Carlsten, C
265
Braun, H
2
Carter, G
302
106,146
Braziel, R.M
349
Casanova, J
11
Choi, S
Brennan, F.H
119
Castan, L
166,43,44,
45,46
142
332
Choo, Q
27
Brenu, E
15,199,74
Castellano, B
Chow, S
Brice, A
133,263
Chow, X
27
Brice, A.M
228
Castiglia, V
202,322,
345
21
79
Castellano, B
Chowdhury, A
29,37
Broadley, S
199
Cavallari, J.F
22
Chu, E
42
Ce, T
275
Chuang, T
232
Cerny, A
75
Chung, Y
233
Brodnicki, T.C
Brooks, A.J
187
14
42
S-7
Campbell, I
Choi, J
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 44
Clare, S
87
Daunt, C
270
Doyon, P
64
Clarke, B.D
234
Davare, M.A
349
Draganidis, D
200
Clarke, D.T
235
Davidson, S
239
Drake, S.L
84
Clerzius, G
34
DAVIS, R
330
Drew, A
251
Clouston, A
188
Day, B
278
Druker, B.J
349
Coccia, E
160
D'Cruz, A
307
Du, J
355
Coll, R.C
237,30
de Kretser, D.M
76
Du, M
201
Collinet, E
75
de Min, C
49,51
Du, P
241
Condon, N.D
347
de Moerloose, P
227
Dua, K
175
Condon, S
193
De Nardo, D
36
Duan, E
264
Conos, S
236
De Valle, E
65
Dubey, P
244
Cons, L
49
De Weerd, N.A
242
Dubey, P.K
299
Conti, B.J
31
de Winher, M.P
13
Dudakov, J.A
351
Conti, B.J
162
S-16
Conzelmann, K
Dungan, L.S
237,30
Cook, A
293,325,98
Dean , M.M
218,240,24
1
99
Dungan, L
148
Duong, F.H
Cook, A.D
109,24
144
75
Decker, T
Durbin, J.E
Cooke, K.R
188
350
327
Deddouche, S
Cooper, M
174
327
Deeg, C
Durbin, R.K
296
200
Durum, S
245
Della Gatta, P.A
14
Dymock, B
85
Denery, S
22
Easton, D
S-15
Denou, E
Ebner, F
21
Edwards, K
164,20,319
Edwards, S
S-16
Edwards, S.C
41
Eickmann, M
80
Eide, C.A
349
Ekert, P
270
Elgass, K
292
Eliam, R
73
Ellyard, J
246
Elowsky, C
157
Elso, C.M
42
Elson, G.C
51
Elyada, E
S-6
Endoh, Y
83
Eo, S
43,44,45,46
De, S
Cornish, A.L
237,260,30,
32
255
Correia, N
245
Deretic, V
S-29
Costa, A
152
Deshpande, P
29,37
Cozijnsen, A
52
Dhagat, U
17,276,78
Crabb, B.S
52
Dhodi, J
37
Cridland, J
338
Diamond, M.S
283
Cridland, S
342
Dickensheets, H
38
Croagh, D
18
Diefenbach, A
122
Croce, C
S-15
Di-Masi, J
6
Croker, B.A
126,290
Dimitriadis, E
243,300
Croker, D.E
260,30,32
Dinarello, C.A
308
Crotta, S
239
Dobson, R
256
Cua, D
S-28
Dodds, S
35
Cumming, H
230,298
Doecke, J
320
Curiel, T.J
35
Doherty, P
S-9
Curiel, T.J
238
Dolton, G
87
Cutler, S.J
320
Dong, C
212
Dagley, L
307
Dong, R
118
Dagley, L.F
33
Donnelly, R
241
Daher, A
34
Donnelly, R.P
38
Esen-Bilgin, N
139,150,
306,312,31
9
86
Dai, J
250
Doran, T
235
Estrada, V
251
Dai, W
S-7
Dottore, M
17,276,78
Etemadi, N
179,47
Dai, Y
107
Dougan, G
87
F. Amado, I
48
Daniel, B.J
238
Dowling, J
149,296
Faddy, H.H
99
Dao, V
35
Dowling, J.K
181,39
Fairfax, K
65
Daubeuf, B
51
Doxastakis, M
S-7
Fallon, P.G
96
Cooper, M.A
Ernst, M
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 45
Fan, H
284
Frankel , G
315
Gavrilescu, N
262
Fancke, B
311
Franklin, B.F
S-14
Gawali, N
37
Fantino, E
6
Franklin, B.S
177
Gearing, J
230
Fatouros, I.G
200
franzen, k
314
Geczy, C
62
Faux, M.C
139
Fraser, F.W
348
Geczy, C.L
79,83
Febbraio, M
S-10
Fraustro, R
84
Geissmann, F
S-30
Felber, B.K
8
Freedman, B.A
220
Gelderblom, M
119
218,219,
240,241
355
Freeman, A.F
220
George, C.X
S-22
Freeman, S
278
Gerlic, M
286
Freitas, A.A
48
Gerondakis, S
312,352
Frenz, T
12
Gersting, S.W
308
Fujimoto, M
178
Geyer, M
177
Feng, D
Feng, J
Ferlin, W.G
Fernandez Ruiz,
D
Ferrando, A.A
49,50,51
245
Fujita, T
500,81
Ghazaryan, H
226
Ferrante, A
248,303
Fullerton, M.D
22
Ghazawi, I
320
Ghosh, G
120
191,52
Ferreira, M
53
Funatake, C.J
221
Ferrero, R
312,93
Fung, K
242,59
Giacomini, E
160
Ffrench, R
311
Fung, K.y
251
Ginhoux, F
S-31
Fickentscher, C
227
Gabay, C
60
Giogha, C
315
Gnatovskiy, L
297
Fink, K
64
Gack, M
307
Finkel, A
230,251
Gad, H
252
Goarant, C
127
Finlay, C
S-16
Gade, P
273
Godfrey, D.I
351
Finlay, C.M
54
Gahan, M.E
248
Godsell, J
324
Gold, M
265
Firmenich, S
105
Gale Jr, M
117
Fischer, K
270
Gale Jr., M
118
Goldberg, G
254
Fish, E.N
317,55
Gale, M.J
113
Goldberg, G.L
20,255,33
Fitzgerald, K
FitzgeraldBocarsly, P
Flach, C
249
Gall, J
152
Golenbock, D.T
336,39
Gallimore, A.M
87
Gomez, G.A
S-7
Gambin, Y
S-7
212,S-11
Gamero, A.M
38
Flavell, R
97
135
Fleming, R
Gamieldien, J
260,32
157
Floudas, C.A
Gangaplara, A
99
298
Flower, R.R
Ganju, V
22
251,352
Foley, K.P
Gantier, M
250
154
Goodall, G
202,322,
332,345
209
Gooley, P
133
Gopalan, B
120
Gorgani, N
303
Gottschalk, G
15
Gough, D
287
Gough, D.J
63
Gonzalez, B
Folkersen,
314
Gantier, M.P
Foo, H
277
Gantier, M.P
176,253,
308
163
Foote, A
262
Gao, X
248
Ford, S
42
Garbi, N
9
Foreman, H
158
Garcia, K
328
Forlino, A
220
Garcia, S
48
García-Sastre, A
326
Gardon, O
S-7
Foster, E.C
230,242,25
1
220
175
340
Foster, P
Garforth, S.J
Griffin, M
118
125
Fowler, K.W
Gargett, C
Garlanda, C
308
Fraitag, S
124
Grigg, J.B
213,256,
319
118
221
188,295
Grigoriadis, G
65
Frank, R.T
Gartlan, K.H
Gatignol, A
34
Grill, M
257,334
Forster, S
Goulet, M
242,251,29
8
116
Gourbeyre, P
14
Grander, D
282
Grandvaux, N
64
Grant, S
107
Gould, J
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 46
Groom, J
130
Haw, T
175
Holland, S.M
220
Groß, C.
165
Hawthorn, A
199,74
Horai, R
331
Gross, C.J
210
Hayashi, H
103
Horne, A
243
Grumont, R.J
312
Hayman, T
307
Horne, W
156
Grywalska, E
258
He, H
106
Horner, S.M
267
Guayasamin, R.C
259
Hearps, A.C
215
Hornung, V
146,S-32
Gugasyan, R
65
Heath, W
191
Horstmann, M
245
Gupta, A
222
Heath, W.R
52
Horvat, J
175
Haddad, C
248
Hedger, M.P
76
Horvath, C.M
229
Hahn, S.A
66
Heikenwälder, M
12
Horvath, G.L
177
Hahtola, S
124
Heim, M.H
75
Hossain, A
133
Hait, N.C
283
Heinen, A.P
105
Hovhannisyan, L
226
Halai, R
260,32
Heinrich, J
148
Hsiang, T
113
Hall, C
286,47
Heinrich, J
77
Hsu, A
175,81,82
Hall, R.A
187
Hellard, M
311
Hsu, K
62,79,83
Hamilton, J
293,325,98
Hellman, A
S-6
Hsu, M.M
84
Hamilton, J.A
109,24,329
Henriksbo, B.D
22
Hsueh, P
26
Hamilton, N.A
277
Hercus, T.R
17,276,78
Hu, J
90
Hams, E
96
Hernandez, P
122
Hu, Y
312
Hamza, H
S-6
Herren, S
51
Huang, C
85
Han Yeo, T
6
Huang, D
120
Huang, D.C
197
Huang, W
283
Huang, Z.H
248
Han, C
67
Han, M
68,69
Han, S
70,71
Hertzog, P.J
Han, Y
43
Heubel, A.D
230,242,
251,298,
S-20
125,181,
184,321
129
Hubel, P
5
Hertzog, P
Haneklaus, M
72
Hibbs, M
65
Huber, A.K
86
Hanieh, H
299
Hibbs, M.L
264
Huber, M
104,80
Hanish, I
175
Hiebert, P
265
Hughes, D
230
Higgins, S.C
237,30
Humphreys, I.R
87
Hii, C
303
Hunter, C
313
Hii, C.S
248
Huntington, N.D
88
Hill, G
295
Hurez, V
238,35
Hill, G.R
188,S-24
Hussan, F
89
Hill, J
342
Huth, T
199,74
Hilton, D.J
S-37
Huynh, J
329
Hirao, I
208
hwang, e
108
Hiroshima, Y
62,79,83
Hyun, J
71
Hirota, J.A
265
I. Hori, J
268
Hixon, J.A
245
Iadonato, S.P
118
Hochdörfer, T
104
Iain, C.L
80
Hochrein, H
266
Ibsen, M
252
59
Hoeksema, M.A
13
Iizasa, E
207
Hartmann, R
252,75
Hoening, S
146
Infusini, G
315
Hassan, M
97
Hofer, M.J
80
Irani, D
86
Hofer, M.J
91
Ireland, D.J
101
51
Hoffmann, H
328
Ireton, R.C
118
124
Hoi, A
324
Irvine, K.M
277
Hannigan, G.E
3
Hanson, C
175,261,81,
82
208
Hara, H
207
Harari, D
73
Hardcastle, S
199
Hardcastle, S.L
74
Hardy, M.P
276,78
Harikumar, K.B
283
Hansbro, P
Harris, J
Harrison, A
Hartland, E
Hartland, E.L
Hasty, P
Hatterer, E
Hautaniemi, S
262,284,
324
133,263
315,S-12
35
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 47
Irving, A
93
irving, a.T
269
Ishida, Y
138
Ito, N
133,228
Iwahashi, M
341
Kadosh, E
100,19,29,
37
S-6
Kaeslin, G
32
Kaiser, L
75
Kakuta, S
331
Kalakonda, S
274
Kalfass, C
148
Juvekar, A
Kho, D
279
Khromykh, A.A
234
Ki, K.K
99
Kieslich, C.A
260
Kile, B
197,278
Kile, B.T
126,301
Kim, B
43
Kim, D
108,110
Kim, H
119
Izumida, M
275,331,35
3
103
Jabbour, A
270
Kalinke, U
148
Jackson, C.J
203
Kalinke, U
12,77
James, R.W
60
Kallfass, C
174
Kim, H
James, S
212
Kallweit, N
73
Kim, I
28,68,69,
S-25
142
Jamurtas, A.Z
200
Kalvakolanu, D
274
Kim, J
43,44,45,46
Jang, D
166
Kalvakolanu, D.V
273
Kim, M
126,68
Kamada, R
500
Kamiya, T
275
Jarnicki, A.G
133,136,
228
139
Kamiyama, H
103
Jaworowski, A
215
S-20
276,78
Jayatilleke, K
Kan, W.L
251
71
Jeffrey, K
Kang, G
161,209
70,71
Jenkins, B
Kang, H
128,16,18
Kang, M
92
Jenkins, B.J
142
Kang, N
70,71
Jeon, H
Kissick, H.T
102
271
Jeong, J
Kang, Y
131,244,29
9
101
Kitching, A.R
42
143
10
Jeong, Y
Kang, Z
S-34
207
Jeskanen, L
Klabackova, S
172
170
93
Jespersen, L
Klenerman, P
87
Jeyaratnam, J
344
Kanneganti, T
KaparakisLiaskos, M
Kapetanovic, R
Kiyohara, H
124
277
Klip, A
23
Ji, S
90
Karagounis, L.G
200
Knight, D
175,265
Jiang,
113
Kastner, D.L
126,30
Ko, H
126
Jiang, Z
S-33
Kato, H
94
Kobarg, J
245
Jin, T
9
Kawaguchi, N
79
Koernig, S
20
John, L
S-22
Kay, E
95
Koh, Y
71
Johnson, F.B
169
Ke, F
301
Kolbe, C.L
177
Johnson, K
346
Kedzierska, K
278
Kolesnik, T
278
Johnson, Z
50
Kedzierski, L
278
Kolesnik, T.B
281,33
Johnston, S
199,74
Kennedy, C
315,59
Kolls, J
156
Jones, E
87
Kennedy, G
188
Kolosenko, I
282
Jones, G.W
128,186
Kershaw, N
307,S-3
Kondo, T
138
Jones, J
139
S-37
Kong, X
283
Jones, S
262,S-13
117
König, K
285
Jones, S.A
186,87
Koo, D
70
Jonuleit, H
66
Kershaw, N.J
Khajornjiraphan,
N
Khan, A.R
272
75
179
Koo, J
Jørgensen, S
Khan, N
271
97
Jung, S
91
Khanna, N
180
Jung, Y
272
Khare, A
156
Kordula, T
Kosco-Vilbois ,
M.H
Köster, M
283
Jung, H
Khan, P
Just, P
221
Khiew, H
98
Köster, M
12
Iwakura, Y
Jans, D.A
96
Kim, T
102,28,280,
43,44,45,46
102,271
Kimoto, M
208
Kimura, A
Kirill Alexandrov,
K
138
Kim, S
Kishimoto, T
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 48
S-7
49,50,51
73
Kostyrko, K
223
Larner, A
107
Lilja, A.R
139
Kotenko, S.V
327,340
Larrous, F
133
Lim, H
233
Lasry, A
S-6
Lim, L.L
255
Lassnig, C
144
Lim, W
27
Lin, A
312
Lin, D
289
Lin, H
26
Lau, L
146,176,
177,237,
296,30,36,
9,S-14
52
Lin, R
116
Lau, M
264
Lin, Y
25,25
Lau, T.C
22
Linder, S
282
Lauterbach, H
266
Lindqvist, L
236
Lawley, T.D
87
Lineburg, K.E
188
Linossi, E
Linossi, E.M
278,281,
307
33
Kovarik, P
100,19,29,
37
21
Koyama, M
188,295
Kreijveld, E
188
Krishnan, B
157,323
Krishnan, S
101
Krishnan, T
243
Krol, I
75
Krug, L
158
Ku, H
102
Kubo, S
353
Lawlor, K
Kubo, Y
103
Lawlor, K.E
193,20,286,
301
290
Kubota, M
207
Lazear, H.M
283
Liongue, C
348
Kueh, A
312
Le Page, M
S-15
Liston, A
48
Kufer, T
93
Leach, J
188
Liu, A
238,35
Kuhny, M
104
Lee, C
101,223
Liu, C
195
Kuninaka, Y
138
Lee, D
142
Liu, H
113
Kuns, R.D
188
Lee, H
102,272
Liu, J
201
Kuprash, D.V
222
Liu, R
290
Kurata, R
353
115,290
Kurschus, F.C
Lee, M
Liu, X
105
108,262,
284,92
109
157
272,71
Liu, Y
Kuszynski, C
Lee, N
Kutalik, Z
75
Lee, S
Ljungberg, L.U
Kvaskoff, D
119
Lo, C
292,93
Kwa, M
329
Lo, C.Y
228,308
Kwak, B.R
60
LeibundGutLandmann, S
Leitner, N.R
110,142,14
2
350
116,232,
331,35
291
144
Lobry, C
287,63
L Zuniga, E
S-35
Leong, D
20
Loh, K
196
Labzin, L.I
36
Leser, G.P
229
Loh, Z
117
Lacey, D
225,293,98
Levin, D
328
Loo, Y
Lacey, D.C
109,24
Levy, D
287,297
Lacroix, M
50
Levy, D.E
63
232
Lewis, R.S
301,348
Lai, S
221
Li, N
67
Lair, D
14
Li, P
168
106
Li, S
308
10
Li, W
91
Lamb, R.A
229
Li, W.Q
245
Lancaster, G.I
S-10
Li, X
10
300
Li, Z
S-22
295
Liau, N
S-3
Lang, T
262,284
Liau, N.P
288
Langley, K.G
S-10
Lienenklaus, S
148
Lao, C
285
Lienenklaus, S
12
Lao, K
27
Lieu, K
133,228,
263
Kothavade, P
Lai, C
Lamaze, C
Lamb, B.T
Lane, N
Lane, S
Latz, E
Lee, J
Lopez, A.F
113,117,
118
17,276,78
Lor, M
188
Lotz, A.S
308
Louis, C
293
Low, J
294
Low, P
119
Loy, J.D
323
Lu, C
232
Lu, T
120
Lubick, K.J
220
Lucet, I.S
33
Ludlow, H
76
Lun, A
191
Luo, C
283
Luo, L
347
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 49
Luther, R.J
48
Marsh, G
MarshallGradisnik, S
Martin, P
235
Mellick, A
320
Luu, K
230
Lynch, J
117
15,199,74
Melo, C
129
Mendoza, J.L
328
Lynch, J.P
196
S-24
Menkhorst, E
243,300
Martinet, L
Lyras, D
302
129
Messmer-Blust, A
344
Martins, J.V
M.S. Pereira, M
268
Ma, J
52
301
Metcalf, D
197,S-37
Mason, K.D
270
Methenitis, S
200
Masouras, D
Ma, Y
303
157,323
Meunier, F.A
119
Massilamany, C
MacDonald, K.P
188,295
313
Michailidis, Y
200
Masters, S
Mackay, F
S-15
MacKenzie, R.J
349
Masters, S.L
Mackin, L
42
Masters, S.L
126,237,30,
36
72
Michelotti, G
S-39
Michiels, T
148
Michonneau, D
124
Magcwebeba, T
121
Masuda, K
244,299
Mielke, L
306,312
Magnan, A
14
Magnus, T
119
Mathew, S
104
Mielke, L.A
130
Matsorakos, G
200
Mildenhall, A
193,301
Mahalingam, S
248,337
Matsui, M
127
Mileto, S
302
Mahlakoiv, T
122
Matsumoto, T
178
Milito, A.D
282
Mahmoudi, S
123
Matsunaga, K
208
Miller, A.L
9
Mailhé, M
48
Matsuyama, T
103
Millrine, D
131
Malayter, D
221
Maxwell, M
65
Mills, K.G
237
Malefyt, R.d
154
Maxwell, M.J
264
Mills, K.H
S-16
Malhotra, A
285
Mazzone, S
196
Mills, K.H
30,41,54
Maliniemi, P
124
McAllister, C.M
S-22
Milstien, S
283
Malinverni, R
75
McArthur, G
320
Mimuro, H
93
Mandalidis, D
200
McArthur, K
197
Min, J
142
Mangan, N
242,309
McAuley, J
296
Minkah, N
158
Mangan, N.E
125
McCabe, T.M
239
Mishra, V
291
Mangia, A
75
McCaskill, J
235
Misiak, A
S-16
Manion, K
217
McClure, B.J
17,276,78
Miyagi, M
120
Manjegowda, S.B
273
McCulloch, D
123
Mizoroki, A
331
McEwan, A.G
277
Mkrtchyan, G
226
McFadden, G.I
52
Mollard, V
52
Mansor, A
149,296,39,
76
89
141
McGinley, A
S-16
Molloy, T
321
Mansor, M
308
McGuinness, N
S-16
Monack, D
S-17
Mantovani, A
119
McKelvey, K
203
Monaghan, K
164
Manzanero, S
10
McKenzie, B
255
Monaghan, P
235
Maphis, N
203
McKenzie, B.S
20
Monk, P.N
260,32
March, L
113
McLeod, L
161
Monks, B.G
237,30
Marcotrigiano, J
64
McLeod, L.E
128
Monnet, E
51
Mariani, M
287,297
McMenamin, P.G
255
Moon, S
142
Marié, I
63
McMillan, N
235
Moore, P
85
Marié, I.
S-7
McNagny, K
265
Moos, S
105
Mark, A.E
188,295
Mean, I
60
Moradpour, D
75
Markey, K.A
S-39
Mechelli, R
160
Morahan, G
42
Markowitz, G
298
Meier, J
107
Morand, E
262,284
Marks, Z
87
Meijerink, J.P
245
Morand, E.F
324
Marsden, M
Meissner, F
146,177
Morikis, D
260,32
Mansell, A
60
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 50
Morita, K
354
Neunlist, M
11,14
O'Donoghue, R.J
139
Moriyama, E
323
Ng, I.H
136
Ogawa, S
353
Morse, C
156
Ng, M
20,254
Okada, K
133
Mortellaro, A
132,189
Ng, S
8
O'Keeffe, M
266,311,42
Morton, C.J
S-7
Ngo, D
305
Okonski, K.M
S-22
Moseley, G
133,263
Nguyen, K.N
340
Oksayan, S
133
Moseley, G.W
228
Nguyen, P
306
Olejnik, A
223
Moujalled, D.M
301
Olver, S.D
188
Mueller, C
2
O'Mara, M
S-7
Muhammad, A
298
Nguyen, T.H
199,242,31
3,74
119
Mukaida, N
26
140,151
Ni, W
Onabajo, O
138
O'Neill, C
Mukaro, V.R
248
278,307
254
Nicholson, S
O'Neill, L
Müller, M
144
281
S-38
Nicholson, S.A
O'Neill, L.A
Müller, W
105
33,S-37
72
Nicholson, S.E
237,30
Müller-Newen, G
Nicola, N
Ong, G
141
Müllhaupt, B
75
Nicola, N.A
243,278,30
7,S-37
281,33
O'Neill, L.A
159
O'Reilly, L
65
Munawara, U
303
Nicos, N
S-3
O'Reilly, L.A
294,312
Muñoz, E.J
118
Nicosia, A
152
Oren, M
S-6
Muñoz-Planillo, R
30
Niiranen, K
124
Oriss, T.B
156
Murakami, M
S-36
Nilsson, S
154
Ortega, J
194
Noble, E
285
Osborne, G
338
Noçon, A.L
257
Ose, T
133
Murphy, J.M
254,255,S3
33
281
Nold, C
231,292
Osorio, F
350
Murray, P.J
174
Nold, C.A
308
Othman, F
89
Mutz, P
135
O'Toole, S
47
Ovaska, K
124
Nachbur, U
304
Oyeniran, C
283
Naderer, T
268
Nold-Petry, C
Ozato, K
500
Nagai, H
231,292,30
9
285,308,32
4
285,309
321
Mvubu, N.E
100,19
Nold-Petry, C.A
324
Paganin, M
245
Nagmoti, D
289
Noor, S.M
348
Pal, A
132
Naik, S
175
Nosaka, M
138
Palmer, G
60
Nair, P
128
Nowell, C
139
Pandeswara, S
238,35
Najdovska, M
178
Nowicka, H
223
Pang, E
254,255,33
Naka, T
331
Nuernberger, C
174
Pang, K
313
Nakajima, A
207
Nunez, G
30,S-18
Pang, S.W
342
Nakama, Y
273,274
Nurieva, R
310
Panousis, C
20
Nallar, S.C
142
Nutt, S.L
24
Papavasiliou, N
251
Nam, J
12
Nyati, K
244
Papenfuss, A.T
42
Namineni, S
119
Nyati, K.K
299
Paquin, A
151
Narayana, V.K
164,20
NYLEN, S
330
Paramel, G
291,314
Nash, A
78
O’Reilly, L.A
301
Park, A
271
Nash, A.D
250
Obata, Y
339
Park, G
158
Natalia, M
13
Obertas, L
310
Park, H
142,265
Neele, A.E
75
O'Carroll, S
279
Park, J
108,143,92
Negro, F
279
Oda, A
353
Park, M
182
Nelson, V
17,276,78
O'Donoghue, R
150
Park, S
142
Nero, T.L
Park, Y
271,272,68,
Murphy, J
Nguyen, T
Nold, M
Nold, M.F
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 51
69
Poh, A.R
139
Reynolds, E.C
329
Parker, B
298,S-20
Pokrovskaja, K
282
Rheinemann, L
174
Parker, B.S
321
Poli, V
158
Ribeiro, D
245
Porter-Gill, P
140,151
Rice, C
328
Preaudet, A
139
Richards, A
210
Parrini, M
17,276,78,
S-7
144
147,81,82
Pribluda, A
S-6
Ricigliano, V
160
Parsons, K
Price, D
Rickard, J
179,47
Pasiarski, M
258
152
199,74
Rieder, M
148
500
140,151
Rinis, N
159
Patel, M
Patel, R.C
343
Price, M.M
ProkuninaOlsson, L
Proudfoot, A
283
Passmore, R
318
Ripley, B
Patil, A
44,45,46
Prow, N.A
187
Ritchie, M
131,244,29
9
197
Pattabhi, S
118
Rizzo, F
160
Robek, M.D
259
Putoczki, T.L
130,213,25
6,294,306,0
00
312
Roberts, A.W
126,S-37
Quach, A
248,303
Robertson, A
296
Quinn, K
152
Robertson, A.A
237,30
Qvist, R
141,153
Robertson, S.J
220
R. Roy, C
268
Rodriguez, E
60
Ra, J
102
Roederer, M
152
Radetskyy, R
34
Rogers, N.c
350
Raffelt, N.C
188
Rolinski, J
258
Raghavan, S
154,194
Romano, S
160
Ragland, R.L
169
Roquilly, A
191
Rahman, T
59
Rose-John, S
Rajasekaran, R.A
157
Ralph, S.J
320
Ramos, S
15,199,74
Ranki, A
124
Rankin, L.C
130
Rao, D
180
Raundhal, M
156
Rautela, J
321,S-20
Raverdeau, M
S-16
Ray, A
156
Ruitenberg, M.J
231,292,30
8,309,324
119
Parker, M.W
Putoczki, T
Pavlakis, G.N
8
Payne, N.L
42
Pearson, J.S
315
Pedersson, J
309
Peel, A
316
Pelka, K
146
Pennell, L.M
317
Peter Wark, P
81,82
Peter Wark, P.P
147
Peters, K
277
Peterson, D
15
Petrek, M
226
Pfaller, C.K
S-22
Pfefferkorn, C
148
Pham, T
87
Phan, M
277
Phesse, T
Phillips, D.J
76
Phillipson, L
305
Philpott, D
93
Rossello, F
161,334,84,
S-21
352
Rossjohn, J
242
Rotimi, L
279
Rotter, B.M
308
Routy, J
34
Rudd, P
117
Rudd, P.A
337
Rudloff, I
Phulphagar, K
117,196,53,
6
146
Pichlmair, A
5
Ray, P
156
Russell, A.P
200
Pieters, R
245
Rayzman, V
20
Ruwanpura, S
161
Piganis, R
251
107
Ruwanpura, S.M
16
Piganis, R.A
181,184
322
Ruzankina, Y
169
Pijnaker, S.M
13
S. Zamboni, D
268
Pikarsky, E
S-6
Raza, A
Recasens Torné,
M
Recasens, M
Sadler, A
205,352
Pillay, B
135
Reddy, J
157,323
Safi, S
141,153
Pillay, M
135
Reddy, P
188
Sagulenko, V
338,342
158
Pimenta, E
218
Reddy, S
Sahoo, A
310
158
Pinar, A
149,296
Reich, N
Salajegheh, A
74
Platanias, L.C
55
Reid, H
242
Saleh, R
325
Poh, A
150
Reis e Sousa, C
350
Salvetti, M
160
Phipps, S
332
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 52
Samarasinghe, T
309
Seo, D
110
Smadbeck, J
260
Samuel, C.E
Sanchez-Aparicio,
M
Sandoval, M.J
S-22
Seo, Y
166
Small, S.H
169
Serada, S
178
Smirnov, S.V
340
Sertori, R
348
Smith, B
313
270
Servant, M
64
Smith, D
60
Sandow, J
208
Sester, D
338,342
Smith, I.M
170
Sanford, M
Sester, D.P
Smith, J
311
Santiago, K.B
31
346
162
Severa, M
160
Smith, P
73
Santiago, K.B
75
Sforcin, J.M
31
Smith, R
147
Santoro, R
252
Sforcin, J.M
162
Smith, T.P
323
Sarkar, S
Shao, F
Smooker, P.M
215
Sarmento, L.M
245
S-23
163
Shariatrian, N
309
Smyth, G
191,254
Sarvestani, S.T
93
SHARMA, S
330
Smyth, G.K
290,312
Sasakawa, C
73
Sharp, P.P
33
Smyth, M
S-24
Sasson, K
Sharp, Z
Smythe, M
171
Sato, H
103
35
277
Shaw, E
34
Snelgrove, R.J
87
Saunders, B.M
164
Shaw, T
224,316
Sobey, C.G
119
Scalzo-Inguanti, K
6
She, Y
120
Song, Y
354
Schagen, J
Sheikh, F
Soubies, S
174
Schembri, M.A
277
38
22
Shen, K
203
Soupe-Gilbert, M
127
Schertzer, J.D
174
Shillling, P
133
Souza, J.G
129
Schilling, M
73
Shimizu, K
331
Souza, S.R
129
Schlapschy, M
Shin, S
Shortman, K
42
Schmeisser, H
4
Souza-FonsecaGuimaraes, F
S-24
Schlegel, K
148
108
Shrivastava, K
Sowder, R
8
146
332
Schmid-Burgk, J
Sieber, O
Spall, S
286,47
36
213
Schmidt, S
Sierecki, E
Spann, K
117,196,6
146
S-7
Schmidt, T
Silke, J
200
329
Spengos, K
Scholz, G.M
Silva, M.C
179,193,28
6,301,47
245
96
328
Sparwasser, T
Schneider, W.M
283
169
Silveira, A.B
245
Spiegel, S
Schoppy, D.W
172,173
350
Silverman, R
205
Splichal, I
Schraml, B.U
172,173
328,73
Silverman, R.H
167
Splichalova, A
Schreiber, G
Sim, J
102
Stacey, K
338,342
277,346
196
Stacey, K.J
Simpson, J
165
Sin, D
265
Stacey, K.J
Schultze, J
165,210,21
6,277,30,33
8
36
87
Schultze, J.L
Sin, W
168
Stacey, M.A
176
87
Schuster, S
Singh, S
250
Stack, G
210
124
95
9
Stadler, R
Sirois, C.M
76
291,314
Staeheli, P
Sebire, K
Sirsjö, A
Seder, R
152
Sjökvist Ottsjo, L
194
Stahl, R
122,148,17
4,333,77
146,177,36
Segal, D
305
Sjökvist-Ottsjö, L
154
Staines, D
15,199,74
Seillet, C
130
Skerra, A
73
Stark, G
120
Semela, D
75
Skuza, E
309
Starkey, M
175
Semmo, N
75
Sly, P
6
Stefanowicz, D
265
Semper, C
144
Sly, P.D
196
Stefanska, A.M
54
Senkevitch, E
245
Sly, W
97
Stirzaker, R.A
126
Schroder, K
Scotland, R.S
326
327
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 53
Stockwell, D
S-37
Tan, K
285
Ullah, M
53
Stomski, F.C
276
Tang, H
335,355
Umetsu, D.T
S-25
Stone, R
219
Tanigawa, M
341
Uno, K
341
Stow, J
347
Tanzer, M
179
Upadhyaya, N
218
Stow, J.L
119
Tarique, M
180
Upham, J.W
196
Uqham, J
117
Ushiki, T
281
Tate, M.D
149,251,27
8,296
181,184,39
Usuwanthim, K
303
Uyangaa, E
43,44,45,46
Vajjhala, P
342
Väkevä, L
124
161
Strawbridge, R
294,301,31
2
314
Strobl, B
144
Taylor, P.R
87
Strugnell, R
194
Taylor, R
220
Stunden, J.H
176
Taylor, S
164
Sturgeon, E
188
Teal, B.E
188
Sturm, A
52
Tedla, N
182,62,79
Stutz, A
177,30
Teh, H
212
Sudo, K
331
S-24
Suen, W
187
75
van Driel, I.R
59
Suhrbier, A
117
van Vliet, C.J
351
Sukkar, M
53
Teng, M.W
Terczyńska-Dyla,
E
Terzis, G
Valhos, R
Van den Bossche,
J
van der Velden, S
Sum, C
168
188
Summers, S
42
Thatcher, E.J
336
Sun, B
90
Thibodeaux, S.R
238
Sun, L
238
Thomas, B
93
SUNDAR, S
330
Thomas, B.J
184,337
Suprunenko, M
80
Thomas, E
6
Van Zyl, A
Vanhaesebroeck,
B
Varelias, A
Vásquez
Sotomayor, F
Vasquez, F
121
Tey, S.K
83
343
Surace, M
Thomas, S.R
Vaughn, L.S
283
258
338
Vaux, D
Surdacka, A
Thygesen, S.J
Suter, M
266
Tiganis, T
351
Vaux, D.L
193,236,28
6,47
301
Sutter, G
12
Ting, S
S-15
Vega-Ramos, J
191
Sutton, C
S-16
Tiquia, R
185
Veldman, A
309
Sutton, C.E
30,41
Tognon, C.E
349
Verma, C
132
Swart, A
121
Tolosa, J.J
147
Vestal, D
344
Swart, P
121
Tong, S
243
Vigano, E
189
Sweet, M.J
216,277
Toribio, M.L
245
Vijayaraj, S
190
Sweet, M.J
165
Toyoshima, S
339
Vilella, A
345
Syme, T.E
334
Tran, E
80
Villacampa, N
202,345
Syme, T.E
257
Tritapoe, J
245
Villadangos, J
S-37
Sze, A
Tacchini-Cottier,
F
Taghavi, N
116
Tsantikos, E
264
Villadangos, J.A
191
Tschopp, J
165
Tsou, W
340
S-22
Tsykin, A
209
Vince, J.E
193,236,28
6
301
Taing, H
17
Tu, X
206
Vincent, F
S-15
Takabe, K
107,283
Tuettenberg, A
66
Vinkemeier, U
S-26
Takahashi, H
212
Tunny, K.A
S-7
Vinuesa, C
501
Takemoto, N
178
Turner, L
93
Vitak, N
346
Talabot-Ayer, D
60
Twohig, J.P
186
Vivian, J
242
Tam, C
S-15
Tyner, J.W
349
Vogel, S.N
38
Tamura, T
500
Uddin, J.M
187
Vogl, C
144
Tan, I.K
42
Ullah, A
117
Wack, A
239
Strasser, A
210
Tate, M
200
Vince, J
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 54
13
13
119
188,295
165
210
Wadi, S
344
West, A
18
Yamamoto, N
103
Waheed, A
97
Whelan, D
133
Yamaoka, S
133
Waisman, A
105
Whelan, D.R
228
Yang, C
25
Wakamatsu, E
339
White, M.J
197,301
Yang, P
S-39
Walduck, A
154
White, S
352
Yang, S
355
Walduck, A.K
194
Whiteford, J.R
95
Yang, Y
166
Wall, A.A
347
Whitney, P.G
350
Yanik, G
188
Wall, S
238
Yao, J
90
Walsh, G
285
Yap, J
300
Walsh, P.T
54
Wicks, I.P
254,255,28
6,305
20,290,33
Walts, A.D
351
103
Wiede, F
Yashima, Y
220
Yeh, D
Wang, B
120
144
232
Wienerroither, S
Yeo, J.C
Wang, D
205,352
194
347
Wijburg, O
Yester, J.W
Wang, H
S-39
118
283
Wilkins, C.R
Wang, L
87
204
Wilkinson, G.W
Yi, M
235,25
Yi, Y
Wang, L.F
269
205,352
110
Williams, B
Yim, H.C
Wang, M
307
163
205
Williams, B.R
Yin, D
Wang, M.L
118
3
206
Williams, B.R
Wang, Q
65
353
Willis, S
Yonezawa, T
206
Yoo, E
Wang, S
195
76
70,71
Wilson, K
Yoon, S
Wang, W
355
330
271
WILSON, M
Yoshida, H
Wang, X
S-39
276,78
207
Wilson, N.J
Wanke, F
133,228
354
Wiltzer, L
Yoshida, Y
105
Yoshizaki, K
Ward, A
123
234
341
Wilusz, J
Young, H.A
Ward, A.C
348
198
208
WInk, D
Yu, C
Waring, P.M
312
243,300
25,301
Winship, A
Wark, P
Watanabe-Smith,
K.M
Waters, M.J
217
S-27
Wither, J.E
Yu, H
175
60
Yu, L
205,209
Woldt, E
168
Yunes, J.A
245
Wong, J
S-7
199,74
Zak, D
152
Wong, N
48
Wong, S
27
Zaker-Tabrizi, L
242
Weaver, C
96
Wong, W
286
Zakharyan, R
226
Weaver, C.T
315
Wood, L
238
Zaman, M
299
Webb, A
33
Woodhead, G
285
Zamoshnikova, A
210
Webb, A.I
S-15
Woodruff, T
32
Zanoni, J.N
129
Wei, A
85
Woodruff, T.M
260
Zenatti, P
245
Wei, F
120
Wright, C.R
200
Zhan, Y
191
Wei, H
313
Wu, R
291
Zhan, Z
115
Weisman, A
80
Xiang, Y
201
Zhang, C
335,355
Weiss, C
Weiss, R
218
Xiao, H
201
Zhang, J
Weiss, S.R
167
Xiao, T
9
Zhang, K
243,281,30
7,S-37
211
Wendeln, A
84
Xie, R
202
Zhang, V
117,196
Weninger, W
S-40
Xu, D
352
Zhang, W
355
Wenzel, S
156
Xue, M
203
Zhang, Y
212,500
Werder, R
117
Yagi, K
341
Zhao, Y
206
Werder, R.B
196
Yamada, A
107,283
Zheng, C
116
Wesoly, J
223
Yamamoto, A
152
Zhong, J
201
349
Wicks, I
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 55
Zhou, J
215
Zimmermann, J
146
Zou, W
238
Zhou, Y
157
Zinger, A
S-6
Zozulya, A
73
Zhu, J
500
Zoon, K.C
4
Zuurbier, L
245
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 56
SPEAKER ABSTRACTS
S-28
IL-23 regulation of innate and adaptive immunity
Daniel Cua
Following the discovery of IL-23-dependent T cell immunity, the past decade has witnessed a major revision of the T-helper subset
paradigm and substantial progress has been made in deciphering the molecular mechanisms of T cell lineage commitment and function.
I will present our recent work on the transcriptional control of TH17 cell development and highlight the protective vs. pathogenic roles of
IL-17 and IL-22 in mucosal tissues. I will also discuss the emerging clinical data showing that antibody-mediated neutralization of IL-17
and IL-23 are remarkably effective for treating immune-mediated inflammatory diseases.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 57
S-29
Immunological manifestations of autophagy
Vojo Deretic1
1. UNM School of Medicine, Albuquerque, Mexico
The broad immunological roles of autophagy span innate and adaptive immunity and are often manifested in inflammatory diseases.
The immune effects of autophagy partially overlap with the hub function of autophagy in metabolism and cytoplasmic quality control but
typically expand further afield to encompass unique immunological adaptations. One of the best-appreciated manifestations of
autophagy is protection against microbial invasion, but this is by no means limited to direct elimination of intracellular pathogens and
includes a stratified array of nearly all principal immunological processes. This presentation will provide a broad background to
immunological roles of autophagy, highlight a few model examples, and report on the newest development demonstrating that the TRIM
family of proteins act as cellular organizers of the core autophagy regulatory machinery and as receptors for selective autophagic
elimination of microbial and endogenous cellular targets.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 58
S-30
Origin, Development and Maintenance of Tissue Resident Macrophages
Frederic Geissmann1
1. King’s College London, London, United Kingdom
Macrophages, Monocyte, and Dendritic Cells (DCs) are myeloid cells, phagocytes, and effectors cells of the innate immune system.
Despite the identification of a number of phenotypic subsets, our understanding of the functions of these cells in vivo remains largely
incomplete. Recent fate-mapping studies from our lab have identified a dual developmental origin for macrophages, monocytes and
DCs. Monocytes, some tissue monocyte/macrophage subsets, such as in the adult gut, classical DCs and plasmacytoid DCs, develop
and renew in vivo from c-Myb-dependent Hematopoietic Stem and Progenitors Cells (HSPCs) and require require the transcription
factor for its development and express Csf1R and Flt3. In contrast a large number of ‘resident’ tissue macrophage networks, including
liver Kuppfer cells, epidermal Langerhans cells, brain microglia, alveolar macrophages, and a proportion of splenic red pulp and kidney
macrophages, develop during embryogenesis and early post-natal life from Csf1R+ Yolk-Sac erythro-myeloid progenitors, that do not
express Flt3- and develop in the absence of c-Myb-. The Yolk-Sac erythro-myeloid progenitors colonize the fetal liver after E9, expand
and give rise to the transient fetal liver hematopoiesis, and to definitive resident macrophage networks. These c-Myb-independent
macrophages persist in adult mice, independently of HSPCs and c-Myb. We propose that differentiation of macrophages should be
studied in the context of the development of macrophage networks.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 59
S-31
Dendritic Cell and Macrophage Ontogeny
Florent Ginhoux1
1. Agency for Science, Technology and Research (A*STAR), ., Singapore
Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
Dendritic cells (DCs), monocytes and macrophages play crucial and distinct roles in tissue homeostasis and immunity, but also
contribute to a broad spectrum of pathologies and are thus attractive therapeutic targets. Potential intervention strategies aiming at
manipulation of these cells will require in-depth insights of their origins and the mechanisms that govern their homeostasis.
DCs and monocytes arise from common bone marrow (BM) precursor named macrophage-dendritic cell precursors (MDP), branching
into exclusively DC- or monocyte-committed progenitors named common dendritic cell progenitors (CDPs) or common monocyte
progenitor (cMoPs) respectively. CDPs give rise to plasmacytoid DC and migratory DC precursors termed pre-DCs. Pre-DCs seed
tissues where they differentiate into the two major functionally specialized DC lineages, CD8α+/CD103+ DCs and CD11b+ DCs.
Recent evidence from our laboratory and others have showed that monocytes do not substantially contribute to all tissue macrophage
populations in steady state and inflammatory conditions. Rather certain tissue macrophages in mice are derived from embryonic
precursors, are seeded before birth and maintain themselves in adults by self-renewal. In addition, we now provide evidence that
commitment to CD8α+/CD103+ DC or CD11b+ DC subsets is imprinted early in the BM. Combining single cell sequencing with
conventional transcriptomic analysis, Cytometry by Time-Of-Flight mass spectrometry (CyTOF) and intra-femoral transfer, we identified
for the first time DC subset-specific precursors in the BM as well as previously unknown molecular checkpoints for DC lineage
commitment as early as the CDP stage.
These new insights into the origins of DCs, monocytes and macrophages should aid the rational design of therapies aimed at
harnessing the functions of these cells in homeostasis and inflammation and will allow efficient targeting and manipulation during health
and disease.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 60
S-32
Cytosolic DNA detection by the inflammasome and beyond
Veit Hornung1
1. University Hospital, University of Bonn, Bonn, Germany
Inflammasomes are large multi-protein complexes that trigger the activation of so called inflammatory caspases, most importantly
caspase-1. Upon activation, caspase-1 cleaves various substrates, including the pro-inflammatory cytokines IL-1β and IL-18, which are
rendered active upon proteolytic maturation. Beyond these members of the IL-1 family, multiple additional caspase-1 targets have been
identified, yet the physiological role of these cleavage events remains to be elucidated. Moreover, caspase-1 activation leads to a
certain type of cell death, named pyroptosis, that can also trigger inflammation in trans. Various sensor proteins have been identified
that can trigger the formation of inflammasome platforms. These inflammasome-forming PRRs, except for the DNA sensor AIM2, belong
to the Nod-like receptor (NLR) family. NLRs are cytosolic PRRs with a tripartite domain architecture comprising of C-terminal leucinerich repeats (LRRs) that are thought to sense microbial molecules or endogenous stress mediators; a central NACHT nucleoside
triphosphatase domain that mediates NLR oligomerization and formation of the core structure of the inflammasome; and an N-terminal
effector domain required for signal transduction. AIM2 is an exception to this rule, as it contains a unique C-terminal ligand binding
domain, the HIN200 domain. While AIM2 plays a non-redundant role in sensing the presence of cytosolic DNA leading to ASCdependent inflammasome activation, other cytosolic DNA sensing mechanisms have been identified that are required to drive proinflammatory gene expression. Among these, the recently identified cGAS/STING axis plays the predominant role. In this talk an update
is given on our recent progress on the characterization of cytosolic DNA sensing pathways and their interconnection in the context of
infectious and sterile inflammatory conditions.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 61
S-33
Identification and Characterization of Phosphodiesterase V-cGAPs That Degrade 3’3’-Cyclic GMPAMP in vibrio cholerae
Zhengfan Jiang
Not available at time of printing
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 62
S-34
IL-1 regulation in inflammatory disease
Thirumala-Devi Kanneganti1
1. Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, United States
Inflammation plays vital roles in protective responses against pathogens and tissue repair, however, improper resolution of inflammatory
networks is centrally involved in the pathogenesis of many acute and chronic diseases. Extensive advances have been made in recent
years to define the inflammatory processes that are required for pathogen clearance, however, in comparison, less is known about the
regulation of inflammation in sterile settings. Our recent studies describe the cellular events and molecular signalling pathways
regulated by the cytosolic innate immune receptors NLRs and IL-1 that govern inflammation in inflammatory disease.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 63
S-35
Dynamics Of Cytokine Responses During Chronic Viral Infection
Elina L Zuniga1
1. University of California, San Diego, San Diego, CAL, United States
Our laboratory is interested in understanding the cellular and molecular mechanisms underlying the regulation of the immune system
during viral infections, particularly infections with persistent viruses, which represent a serious health problem. To overcome the lack of
small animal hosts for human persistent viruses, we use models of chronic viral infections in their natural murine hosts. In particular, by
using chronic lymphocytic choriomeningitis virus (LCMV) infection in mice we have evaluated the dynamics of several cytokines
(including interleukin-6 and transforming growth factor-b) and investigated their regulatory function on immune cells and their impact on
the outcome of the infection at different times after viral challenge. By doing in-vivo studies at the whole organism level we are able to
assess the impact of selected cytokine signaling in the context of the persistent infectious environment in which the immune cells are
navigating. I will be presenting recent discoveries in this research area.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 64
S-36
Masaaki Murakami1
1. Hokkaido University, Japan
Our laboratory is interested in understanding the cellular and molecular mechanisms underlying the regulation of the immune system
during viral infections, particularly infections with persistent viruses, which represent a serious health problem. To overcome the lack of
small animal hosts for human persistent viruses, we use models of chronic viral infections in their natural murine hosts. In particular, by
using chronic lymphocytic choriomeningitis virus (LCMV) infection in mice we have evaluated the dynamics of several cytokines
(including interleukin-6 and transforming growth factor-b) and investigated their regulatory function on immune cells and their impact on
the outcome of the infection at different times after viral challenge. By doing in-vivo studies at the whole organism level we are able to
assess the impact of selected cytokine signaling in the context of the persistent infectious environment in which the immune cells are
navigating. I will be presenting recent discoveries in this research area.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 65
S-37
Old and new families of negative regulators of cytokine signalling
Nicos Nicola1, Donald Metcalf1, Warren S Alexander1, Douglas J Hilton1, Sandra E Nicholson1, Jian-Guo Zhang1, Andrew W
Roberts1, Alan Ching1, Jose Villadangos1, Dina Stockwell1, Nadia J Kershaw1, Jeff J Babon1
1. Walter and Eliza Hall Institute, Parkville, VIC, Australia
Our discovery of a family of negative regulators of cytokine signalling (the Suppressors of Cytokine Signalling, SOCS) was based on a
screen that allowed the M1 myeloid leukaemic cell line to continue to grow in the presence of interleukin 6.
The expression of various SOCS proteins is induced by cytokine activation of the JAK/STAT pathway and they then act as feedback
inhibitors to terminate the pathway. Biochemical studies have revealed that the SH2 domains of SOCS proteins bind phosphorylated
tyrosines on selected substrates and that the C-terminal SOCS box recruits elonginsB/C and cullin 5 to form an E3 ubiquitin ligase that
ubiquitinates the bound substrates and targets them for proteasomal degradation.
Two members of the family, SOCS1 and SOCS3, display an additional inhibitory mechanism that involves direct inhibition of the kinase
activity of JAKs. Recent structural and biochemical studies have revealed the precise mechanism by which this occurs. Genetic deletion
studies in mice have revealed how overactivity of cytokine action and specificity of action are regulated by SOCS proteins and how
these relate to the molecular mechanism of action. Very recently we have used the same M1 assay described above to discover a
second family of inhibitors of cytokine signalling- the MARCH proteins. These proteins are integral membrane proteins that also display
E3 ubiquitin ligase activity and have been previously shown to regulate the expression at the cell surface of a number of
immunoregulatory receptors. We now show that some of these proteins also specifically down-regulate the cell surface expression of
cytokine receptors like those for interleukin 6 and render cells unresponsive to the cytokine.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 66
S-38
A lifelong love affair with IL-1 and inflammation
Luke O'Neill1
1. Trinity College Dublin, Dublin, Ireland
IL-1 was one of the first cytokines to be described, first as a bioactivity that mediated inflammation in many different disease contexts,
and then as a family of proteins – IL-1alpha, IL-1beta and the IL1 receptor antagonist. By the late 1980s IL-1 was known to induce the
expression of a large number of immune and inflammatory genes and a key mystery was how it signals. Great detail into the signalling
pathways leading to NF-kappaB and p38 MAP kinase then emerged. The IL-1 receptor was the founder member of the IL-1R/TLR
superfamily and was first defined in detail 15 years ago. Since then, there has been remarkable progress in our understanding of both
branches of the superfamily. Ligands have been described for most receptors. Within the IL-1R subfamily, notable examples include
IL33 for ST2 and IL-36 for IL-1Rrp2.
The role of TLRs in the sensing of microbial products led to a renaissance of interest in innate immune mechanisms and represented
one of the biggest advances in our understanding of the immune system over the past 50 years. For investigators interested in signal
transduction, the area has proved very fruitful in terms of the discovery of new signalling pathways and processes, notably the MyD88
family of adapter proteins and the IRAK family, involving a complex series of ubiquitination and phosphorylation reactions.
Studies on how the-pro form of IL-1beta is processed led the description of inflammasomes, multiprotein complexes that activate
caspase-1. Inflammasomes can also trigger a type of cell death dubbed pyroptosis. The best understood inflammasome comprises
Nlrp3, and this protein has been implicated in all of the pathologies suggested to involve IL-1 in the 1980s which even then included
gout, atherosclerosis, Alzheimer’s disease and Type 2 diabetes.
We therefore have a love story beginning as an ill-defined pro-inflammatory factor in supernatants from LPS-activated macrophags, to a
revolution in innate immunity, and the real prospect of limiting the IL-1 system for substantial clinical benefit.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 67
S-39
The Paradox roles of IL-18 Signaling in Hepatocellular Carcinoma
Xiao-Fan Wang1, Geoffrey Markowitz1, Pengyuan Yang1, Gregory Michelotti1, Hongyang Wang1
1. Duke University Medical Center, Durham, NC, United States
Hepatocellular carcinoma (HCC) is the fifth most common cancer and third leading cause of cancer-related mortality world-wide. This
cancer almost always develops upon the background of chronic pathological inflammation and resultant fibrosis, which regulate both the
development and progression of this cancer. Interleukin 18 (IL-18), a member of the IL-1 family of cytokines, has been reported to be
elevated in serum of patients with liver disease and HCC, however, its mechanistic significance remains poorly understood.
To explore this question, we evaluated patient serum and tissue samples, and found that while IL-18 was indeed elevated in comparison
to healthy individuals, and its elevation correlated to worse survival of patients, its levels were decreased inside tumor compared to nontumor tissue from the same patient. Utilizing an immune-competent mouse model with orthotopic tumor implantation into a fibrotic liver,
we found that both knocking down IL-18 expression in mouse HCC cells implanted into wild-type mice and inoculating mouse HCC cells
in an IL-18 receptor-deficient mouse resulted in enhanced tumor growth. Post-implantation, CD8+ T-cells were reduced and CD4+ Tcells mildly increased in tumors in the IL-18 receptor-deficient mice compared to wild-type controls, with a resultant robust change in
CD8:CD4 T-cell ratio, while other IL-18-responsive cell types, such as NK cells, were unchanged. A chemically-induced carcinogenesis
model also displayed increased tumor burden in IL-18 receptor-deficient mice compared to wild-type controls. Re-examination of patient
samples revealed that magnitude of the difference in IL-18 expression between tumor and non-tumor liver correlated with patient
prognosis, with worse prognosis in patients with less IL-18 inside the tumor compared with non-tumor liver tissues. These results
suggest a possible biphasic role for IL-18 in the pathological process of this disease, and demonstrate a complex role for IL-18 in HCC
tumorigenesis and progression.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 68
S-40
The role of perivascular macrophages in neutrophil recruitment into the skin
Wolfgang Weninger1
1. Centenary Institute, Camperdown, NSW, Australia
Neutrophils are important defenders against bacterial infections. Upon bacterial entry into the skin, neutrophils migrate rapidly into the
inflamed dermis. We found that perivascular macrophages (PVM) are essential regulators of neutrophil homing following infection with
the pathogen Staphylococcus aureus (S. aureus). PVM are juxtaposed to post-capillary venules in the dermis, and are capable of taking
up molecules from plasma. Using real-time multiphoton intravital microscopy, we show that neutrophils extravasate from inflamed
venules in close contact with PVM. The latter are a major source of the neutrophil chemo-attractants CXCL1 and 2. Strikingly, the S.
aureus virulence factor alpha-hemolysin targets macrophages, resulting in their lysis. Consequently, neutrophil recruitment is delayed
leading to increased bacterial survival and tissue necrosis. We further demonstrate that macrophages express high levels of the
hemolysin receptor ADAM-10 and that pharmacologic blocking of ADAM-10 interferes with macrophage killing by hemolysin in vitro. In
summary, we reveal the thus far unknown guidance of blood-borne cells by perivascular cells resulting in extravasation. We propose the
existence of an integrated multi-cellular unit, termed the perivascular extravasation unit (PVEU), which, besides endothelial cells and
pericytes, includes PVM. Together, PVEU components regulate leukocyte recruitment into the skin. Finally, the killing of PVM by a
bacterial product provides insight into the mechanisms of immuno-evasion by a human pathogen.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 69
MEETING ABSTRACTS
213
Characterisation of the function of the Interleukin-11 signaling complex in human disease
Suad Abdirahman1, Oliver Sieber1, Michael Griffin2, Tracy Putoczki1
1. Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
2. Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne,
Melbourne, VIC, Australia
Interleukin (IL)-11 is an under-characterised member of the IL-6 family of cytokines that signals through a cell-type specific a-subunit
receptor, IL-11Ra, and a ubiquitously expressed transmembrane b-subunit receptor, GP130. The formation of the active hexameric IL11/IL-11Ra/GP130 (2:2:2) signaling complex leads to the recruitment of intracellular Janus Kinases (JAKs) which phosphorylate the
cytoplasmic tail of GP130 providing docking sites for the signal transducer and activator of transcription (STAT) proteins. Once
phosphorylated, STATs form active dimers that translocate to the nucleus where they regulate the expression of numerous genes
involved in cell survival and proliferation. Recently, mutations in components of the IL-11 signaling complex have been linked to a range
of human disorders including craniosynostosis.
Our current understanding of the structural mechanism of IL-11 signaling has been based on homology models using IL-6, despite low
sequence similarity (~20%). IL-6, signals through a hexameric IL-6/IL-6Ra/GP130 (2:2:2) signaling complex, for which high resolution
crystal structures are available. We have recently solved the first crystal structure for human IL-11, which suggests important differences
between these two cytokines including how they engage GP130 and trigger the downstream signaling cascade. In order to understand
how mutations in components of the IL-11 signaling complex identified in human disease impact on signal transduction, we have
performed site-directed mutagenesis of key residues at the IL-11/IL-11Ra and IL-11Ra/GP130 interface. We characterised the impact of
each mutation on the phosphorylation of STAT proteins in BA/F3 cells, which otherwise lack IL-11/IL-11Ra/GP130 components. Our
results identify regions of the IL-11 signaling complex that are crucial for signaling, and represent druggable target sites to agonise or
antagonise this signaling pathway.
215
HIV infection is associated with altered monocyte subset basal cytokine levels and responsiveness
to LPS stimulation.
Thomas A Angelovich1, 2, Jingling Zhou2, Peter M Smooker1, Anna C Hearps2, 3, Anthony Jaworowski4, 2, 3
1. School of Applied Sciences, RMIT University, Melbourne, Victoria, Australia
2. Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia
3. Department of Infectious Disease, Monash University, Melbourne , Victoria, Australia
4. Department of Immunology, Monash University, Melbourne, Victoria, Australia
Introduction: HIV+ individuals are at increased risk of inflammatory age-related disease despite effective suppression of viremia with
combination antiretroviral therapy (cART). Lipopolysaccharide (LPS) is elevated in these individuals and stimulates monocytes via the
Toll-like receptor 4 pathway producing IL-6 and TNF. We hypothesise that chronic LPS exposure enhances monocyte response to LPS
in HIV+ individuals that exacerbates chronic inflammation and the risk of developing age-related disease.
Methods: The inflammatory state of the three monocyte subsets (classical, intermediate and non-classical) from cART naïve, viremic
(n=10) and virologically supressed (VS) HIV+ donors (n=10) was evaluated by measuring basal and LPS-induced (10 ng/mL, 4 h) levels
of IL-6 and TNF by whole blood intracellular cytokine staining via flow cytometry and mRNA gene expression and results compared to
age-matched HIV- controls (n=10). Soluble signalling components (LPS, LBP), inflammatory markers (CXCL-10) and surface TLR-4
were also measured.
Results: All monocyte subsets from viremic and VS HIV+ individuals showed increased basal levels of intracellular IL-6 and TNF in
comparison to HIV- individuals (p<0.001 for all comparisons). Furthermore, these groups showed enhanced IL-6 and TNF production
when stimulated with LPS (p<0.05 for all comparisons). While plasma LPS and LBP levels were elevated in HIV+ individuals in
comparison to controls (p<0.05 for both), mixing serum from HIV+ individuals with control monocytes did not enhance response to LPS
challenge ex vivo suggesting enhanced monocyte response to LPS in HIV+ individuals is governed by mechanisms intrinsic to the
monocyte. This is supported by increased surface TLR-4 expression on all monocytes subsets for viremic HIV+ individuals (p<0.05).
Conclusions: Monocytes from HIV+ individuals, regardless of therapy status, show increased basal intracellular IL-6 and TNF levels
and a heightened response to LPS which may contribute to chronic inflammation and related inflammatory diseases.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 70
216
Characterization of human-restricted TLR4 responses identifies a novel ubiquitin ligase required for
NLRP3 inflammasome responses in human macrophages
Juliana K Ariffin1, Kate Schroder1, Matt J Sweet1
1. The Institute for Molecular Bioscience, Brisbane, QLD, Australia
We previously characterized conservation and divergence in Toll-like Receptor (TLR)4-dependent transcriptional responses in human
versus mouse macrophages. In this study, we focused on orthologous human and mouse genes uniquely regulated by
lipopolysaccharide (LPS) in primary human macrophages. We confirmed “human-specific” LPS regulation of these genes by examining
expression in primary human macrophages versus a panel of different mouse macrophage populations. In investigating potential
functions, we found that one of these genes (a novel ubiquitin ligase) was required for NLRP3 inflammasome responses, as
demonstrated by gene knock-down in both primary human macrophages and PMA-differentiated THP-1 cells. The NLRP3
inflammasome is a multi-protein complex containing NLRP3, ASC and caspase-1. Activation of the NLRP3 inflammasome results in the
processing and release of the pro-inflammatory cytokines IL-1beta and IL-18, as well as pyroptotic cell death. NLRP3 assembly requires
both a priming signal such as that delivered by the TLR4 agonist LPS, as well as a triggering signal such as damage-associated host
molecules (e.g. ATP) or pathogen-associated stimuli. Recent studies have demonstrated that direct deubiquitination of NLRP3 is
required for triggering of the NLRP3 inflammasome. Our findings now suggest that one of the mechanisms by which LPS primes
inflammasome responses in human macrophages is via the upregulation of a specific ubiquitin ligase. We provide evidence that this
ubiquitin ligase affects LPS priming of inflammasomes in human macrophages by promoting ubiquitination-dependent degradation of
p21(Waf1/Cip1), which has previously been shown to inhibit LPS-inducible IL-1beta expression independently of its cell-cycle inhibitory
role. Current studies are aimed at further exploring the dynamics of p21 protein regulation by the ubiquitin ligase, as well as p21
regulation of IL-1beta synthesis. These findings provide new insights into mechanisms affecting pathological inflammatory processes
and provide direction for potential treatment strategies.
217
+
IL-10 is critical for the expansion of peritoneal and splenic CD5 B cells
Yuriy Baglaenko1, 2, Kieran Manion1, 2, Nan-Hua Chang2, Joan E Wither1, 2
1. University of Toronto, Toronto, Canada
2. Genetics and Development, University Health Network, Toronto, Canada
Aims: Interleukin-10 (IL-10) is a pleiotropic cytokine with immunosuppressive properties that has been shown to support B cell survival.
Recent studies have identified a subset of regulatory B cells that can suppress the progression of autoimmunity by production of IL-10.
Previous work from our laboratory has shown that the introgression of a chromosome 4 interval spanning 32 to 150Mb from
autoimmune prone New Zealand Black mice expands two innate-like lymphocyte populations, CD5+ B cells and Natural Killer T cells.
Additional work on these mice identified the expanded splenic and peritoneal CD5+ B cell population as highly IL-10 competent and
regulatory in nature. The aim of this study was to identify a possible role of IL-10 in the development or homeostatic expansion of NKT
and CD5+ B cells.
Methods: IL-10 knockout mice, obtained from Jackson laboratories, were bred onto our chromosome 4 congenic background. Congenic
knockout mice and controls were aged to 18 - 23 weeks. Splenic and peritoneal cell populations were identified de novo by flow
cytometry.
Results: Similar to previous studies, the IL-10 knockout reduced the proportion of splenic Natural Killer cells on both the B6 and
chromosome 4 backgrounds when compared to IL-10 sufficient controls. Interestingly, IL-10 knockout had no impact on the expansion
of splenic NKT cells, suggesting that IL-10 is not important in the expansion of this population. Surprisingly, IL-10 knockout prevented
the expansion of both splenic and peritoneal CD5+ B cells in chromosome 4 congenic mice and greatly expanded the splenic marginal
zone compartment.
Conclusions: Taken together, these data indicate that IL-10 may play a unique and critical role in sustaining the genetic expansion of
CD5+ B cells in the splenic and peritoneal compartments. Unlike previous reports, this study suggests that the expansion of regulatory B
cells may be impacted by the loss of IL-10.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 71
218
Intrinsic and extrinsic mechanisms of metastatic inhibition by IRF5 in human ductal carcinoma
Betsy Barnes1, Erica Pimenta1, Ryan Weiss1, Saurav De1, Di Feng2, Neelam Upadhyaya1
1. Rutgers Biomedical and Health Sciences, Newark, NJ, United States
2. Boehringer Ingelheim, Ridgefield
Metastasis of primary breast cancer to distant sites and recurrence to incurable disease are the main causes of breast cancer fatalities.
While migration of breast cancer cells out of a duct or lobule is a prerequisite for invasion and metastasis, the ability of these cells to
migrate at all is due to intrinsic intratumoral and extrinsic microenvironment changes. Little is known of the factors that regulate these
intrinsic and extrinsic functions. Previous work has shown that expression of the transcription factor interferon regulatory factor 5 (IRF5)
is significantly decreased as a breast lesion progresses from a non-malignant stage to ductal carcinoma in situ and is eventually lost in
~80% of invasive ductal carcinomas examined. Human in vitro and murine in vivo models of invasive breast cancer cell growth confirm
an important role for IRF5 in regulating breast cancer cell migration, invasion and metastasis. We recently identified the protein domain
necessary for the intrinsic migratory function of IRF5 and found that this function is cytoplasmic and transcription-independent. Given
that IRF5 is also a key transcriptional immune regulator, we examined cytokine/chemokine expression in IRF5(+) and (-) breast cancers
grown in 3D culture and found that IRF5 positively regulates the expression of key cytokines/chemokines, such as CXCL13, that define
a pro- or anti-tumor immune microenvironment. We found that anti-tumorigenic primary T cell subsets, including Th1, Treg and Tfh cells,
were specifically recruited to IRF5(+) tumors as compared with IRF5(-) tumors. We also found that CD19+CXCR5+ B cells were
specifically recruited to IRF5(+) tumors. This finding is intriguing since CXCR5+ B and T cells (Tfh) are essential for the formation of
tertiary lymphoid structures (TLS). Together, these data support that IRF5 controls intrinsic mammary epithelial cell migration and
directly regulates a network of genes that shapes a tumor immune response.
219
Development of new automated technology for the detection and analysis of pathogenic
autoantibodies in autoimmune disease
Betsy Barnes1, Di Feng2, Rivka Stone3
1. Rutgers Biomedical and Health Sciences, Newark, NJ, United States
2. Boehringer Ingelheim, Ridgefield
3. University of Miami, Miami
Anti-nuclear antibodies (ANA) are considered a hallmark of autoimmune rheumatic diseases. An ANA test is routinely performed on
serum from patients presenting with symptoms of autoimmune diseases such as systemic lupus erythematosus (SLE), autoimmune
hepatitis, rheumatoid arthritis, polymyositis/dermatomyositis, mixed connective tissue disease, Sjögren’s syndrome, and systemic
sclerosis. The standard method for ANA detection is the indirect immunofluorescence (IIF) assay on human epithelial-2 (HEp-2) cells
(ANA-HEp-2 test). This assay is a slide-based microscopic analysis that allows for the detection of clinically relevant autoantibodies
generated against cytoplasmic and nuclear antigens in patient serum. One of the major issues with the ANA-HEp-2 test lies in the
subjective evaluation of HEp-2 slides that complicates standardization and reproducibility. We have developed a new high-throughput
process of ANA testing that eliminates the subjectivity of data interpretation, and capitalizes on the properties of imaging flow cytometry
to accurately localize, detect and quantify multiple autoantibodies in a single run. The method was initially developed using anti-nuclear
antibody (ANA) reference sera, prepared by the Arthritis Foundation and the Centers for Disease Control, that contains nuclear
components useful in the diagnosis and classification of autoimmune diseases. Samples were run on an Amnis Imagestream and
algorithms that distinguish between individual staining patterns using masks and features provided in Amnis IDEAS® software were
generated. The method was then tested using complex sera from patients with SLE and data compared to the clinical standard ANAHEp-2 test. We obtained 100% replication of the standard ANA-HEp-2 assay with much greater sensitivity and detection of potentially
new pathogenic autoantibodies. The detection of such new and known autoantibodies is of significant clinical diagnostic value since it
allows for not only the identification of severe autoimmune disorders but also helps to make predictions about their course and
prognosis.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 72
220
Viral antagonism of type I interferon responses reveals prolidase as a regulator of IFNAR1 trafficking
and expression.
Sonja M Best1, Kirk J Lubick1, Shelly J Robertson1, Brett A Freedman1, R. Travis Taylor2, Erin C Foster1, Abhilash I Chiramel1,
Steven M Holland3, Antonella Forlino4, Avram D Walts5, Alexandra F Freeman3, Manfred Boehm5
1. NIAID/NIH, Hamilton, MT, United States
2. University of Toledo School of Medicine, Toledo, OH, USA
3. NIAID/NIH, Bethesda, MD, USA
4. University of Pavia, Pavia, Italy
5. NHLBI, Bethesda, MD, USA
Flaviviruses, including tick-borne encephalitis virus (TBEV), West Nile virus (WNV) and dengue virus, cause severe disease in humans
on a global scale. All flaviviruses antagonize type I interferon (IFN-I)-dependent activation of the JAK-STAT signaling cascade,
suggesting that this pathway is an essential target for flavivirus evasion of innate immunity. However, despite nearly 10 years of
research, the molecular mechanism of inhibition utilized by TBEV and WNV is largely unknown. IFN-I signals through two receptor
subunits, IFNAR1 and IFNAR2. TBEV and WNV antagonize IFN-I responses at a point proximal to the receptor as all signaling events
downstream of this are inhibited in virus-infected cells. Replication of WNV (strain NY99) is associated with degradation of IFNAR1 via
lysosomes, implicating IFNAR1 as the target of viral interference (Evans et al. 2011. Viral Immunol. 24:253). Here we show that TBEV
and WNV downregulate IFNAR1 expression at the protein level. The viral protein responsible is NS5, the major flavivirus IFN-I
antagonist. We show that NS5-mediated loss of IFNAR1 is dependent on binding to a host cell dipeptidase called prolidase (PEPD).
Knockdown of PEPD expression prevented IFNAR1 maturation (glycosylation), increased the ubiquitination of IFNAR1, and promoted
degradation of immature (partially glycosylated) IFNAR1 via the lysosome. In humans, genetic prolidase deficiency (PD) is associated
with recurrent infections although the reasons for immune dysfunction are not known. Our results suggest that IFN-I signaling may be
compromised, which was confirmed in primary fibroblasts from PD patients that expressed low IFNAR1 associated with reduced
STAT1/2 phosphorylation and ISG expression following IFNβ stimulation. Thus, by understanding flavivirus antagonism of IFN-I
signaling, we have uncovered PEPD as a central regulator of cellular responses to IFN-I humans and newly identified PD as a primary
immune deficiency. Manipulation of IFNAR1 stability by therapeutic targeting of PEPD may represent new treatments for both viral and
autoimmune diseases.
221
Correlation and kinetics of IFN-gamma and TNF-alpha transcription and translation in lymphocyte
subsets using the FlowRNA assay and intracellular antibody staining
Richardk T Frank1, Steve Lai1, Adam Best1, Dylan Malayter1, Castle J Funatake1, Peggy Just1
1. eBioscience, Affymetrix , San Diego, CA, USA
Aims:
Intracellular staining and flow cytometric analysis of lymphocytes is commonly used to assess cytokine production at the single-cell level
in heterogeneous samples. Here, we describe a novel fluorescence in situ hybridization (FISH)-based flow cytometry assay (PrimeFlow
RNA Assay) used in combination with intracellular antibody staining to study the kinetics of the transcription and translation of IFNgamma and TNF-alpha in lymphocytes.
Methods:
Normal human peripheral blood mononuclear cells were stimulated with PMA and Ionomycin for 0-5 hours. Cells were fixed,
permeabilized, and intracellularly stained with antibodies for CD8, IFN-gamma, and TNF-alpha. Next, cells underwent a series of
hybridization steps to label mRNA for IFN-gamma and TNF-alpha. Data were collected on an LSRFortessa and analyzed using FlowJo
software.
Results:
IFN-gamma mRNA was upregulated in CD8+ and CD8- lymphocytes within 1 hour after stimulation, while protein levels were not
detected until 2 hours, after which both mRNA and protein were maintained for the next 3-4 hours. In contrast, TNF-alpha mRNA and
protein were both upregulated within 1 hour after stimulation and expression was maintained in CD8+ cells while expression in CD8cells peaked between 1-2 hours and then decreased over the next 4 hours, with the decrease in mRNA preceding the decrease in
protein.
Conclusions
Using the PrimeFlow RNA Assay, we found that induction of IFN-gamma and TNF-alpha mRNA and protein exhibit unique kinetics and
that TNF-alpha protein and mRNA are differentially regulated in CD8+ and CD8- lymphocytes. This new PrimeFlow RNA Assay enables
the study of gene expression at the single-cell level in heterogeneous samples without the need for sorting specific subsets and the
ability to compare and contrast the kinetics of mRNA and protein induction.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 73
222
Autocrine CXCL13-CXCR5 signaling potently induces EMTof breast cancer cells via RANKL-SrcPI3Kp110α during lymph node metastasis
Subir Biswas1, Arnab Gupta2, Dmitry V Kuprash3, Arindam Bhattacharyya1
1. Immunology Lab, Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
2. Department of Surgical Oncology, Saroj Gupta Cancer Centre and Research Institute, Kolkata, West Bengal, India
3. Laboratory of Immunoregulation, Engelhardt Institute of Molecular Biology, Moscow, Russia
Background
Epithelial to mesenchymal transition (EMT) is an important hallmark of metastasis. Increased EMT is associated with lymph node
metastasis (LNM). We investigated EMT inducing potential of chemokine CXCL13 in breast cancer (BC) cell lines and CXCL13-CXCR5
signaling in primary breast tumors in association with different epithelial and mesenchymal markers and EMT inducers.
Methods
Chemokine CXCL13 and and its sole receptor CXCR5 were evaluated in 98 primary breast tumor samples from breast cancer patients
with infiltrating duct carcinoma (IDC). BC cell lines were transduced with CXCR5, treated with recombinant CXCL13 and assessed for
cell migration, EMT markers and activation of intracellular signaling pathways.
Results
A total of 56 patients were LNM positive and 42 were LNM negative. LNM positivity was positively associated with co-expression of
CXCL13 and CXCR5. Increased expression of various mesenchymal molecular markers and regulators and decreased expression of
epithelial marker E-cadherin was found in CXCL13-stimulated BC cells and tumors from CXCL13-CXCR5 co-expressing patients.
Experiments with protein kinase inhibitors demonstrated that CXCL13 stimulates EMT and expression of matrix metalloproteinase
(MMP9) via RANKL-Src axis.
Conclusion
This study demonstrated the prognostic value of CXCL13-CXCR5 co-expression in primary BC. Moreover, it showed the EMT inducing
potential of CXCL13. RANKL-Src axis may present a therapeutic target in LNM positive BC patients.
223
IFNɑ engages a sustained antiviral response depending on STAT2/IRF9 but not ISGF3
Kasia Blaszczyk1, Adam Olejnik1, Yi Ling Chen2, Stefan Chmielewski1, Kaja Kostyrko1, Hanna Nowicka1, Joanna Wesoly1,
Chien-Kuo Lee2, Hans Bluijssen1
1. Department of Human Molecular Genetics, A. Mickiewicz University, Poznan, Poland
2. Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
Evidence is accumulating for the existence of a STAT2/IRF9-dependent, STAT1-independent IFNα signaling pathway. However, no
detailed insight exists in the genome-wide transcriptional regulation and the biological implications of STAT2/IRF9 dependent IFNα
signaling as compared to ISGF3. In STAT1 KO cells overexpressing STAT2 we observed that the IFNα-induced expression of classical
interferon stimulated genes (ISGs) such as human OAS2 and mouse Ifit1 correlated with the kinetics of STAT2 phosphorylation, and the
presence of a STAT2/IRF9 complex requiring STAT2 phosphorylation and the STAT2 transactivation domain.
Subsequent microarray analysis of IFNα treated WT and STAT1 KO cells over-expressing STAT2 extended our observations and
identified around 120 known antiviral ISRE-containing ISGs commonly up-regulated by STAT2/IRF9 and ISGF3. The STAT2/IRF9
directed expression profile of these ISGs was prolonged as compared to the early and transient response mediated by ISGF3. ChIP-seq
analysis confirmed binding of STAT2 to the promoter of a selection of commonly up-regulated ISGs, in an IFNα-dependent manner in
the absence of STAT1.
In addition, we identified a group of “STAT2/IRF9-specific” ISGs, whose response to IFNα was ISGF3-independent. Finally,
STAT2/IRF9 was able to generate a sustained antiviral response upon EMCV and VSV.
Our results further prove that IFNα-activated STAT2/IRF9 induces a prolonged ISGF3-like transcriptome and generates an antiviral
response in the absence of STAT1. Moreover, the existence of “STAT2/IRF9-specific” target genes predicts a novel role of STAT2 in
IFNα signaling.
Possibly, in cells with prolonged STAT2 phosphorylation kinetics this could provide a level of redundancy to ensure effective induction of
an antiviral state and be beneficial for example against viruses that directly block STAT1 and impair the formation of ISGF3.
Preliminary results imply that this situation is very likely to be cell-type specific, where both complexes seem to be involved in different
stages of the antiviral response; ISGF3 stimulating a rapid and transient antiviral response whereas STAT2/IRF9 being responsible for a
more prolonged antiviral response.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 74
224
The rs12971860 C/T SNP in the interferon lambda3 (IL28B) gene influences the expression of
thymidine phosphoryase and cytidine deaminase, two enzymatic markers of inflammation
Sara Bonanzinga3, 2, 1, Tim Shaw2, 4
1. Molecular Microbiology, Victorian Infectious Diseases Reference Laboratory, Melbourne, VIC, Australia
2. Peter Doherty Institute, Melbourne, VIC, Australia
3. Department of Microbiology, Monash University, Clayton, VIC, Australia
4. Molecular Pharmacology, Victorian Infectious Diseases Reference Laboratory, Melbourne, VIC, Australia
Background
Some chronic viral infections are responsive to treatment with interferon alpha (IFN-a), but outcomes are variable and unpredictable. In
the case of chronic hepatitis C virus (HCV) infection, single nucleotide polymorphisms (SNPs) affecting the gene for interferon IFN-L3,
(alias interleukin-28B (IL28B), affect response to IFN-a-based treatments. The most studied SNP is C/T at rs12971860 in the IFN-L3
gene’s CpG rich promoter region, with the ranked probability of favourable response (viral clearance) being CC>CT>TT. The biological
mechanisms responsible for this phenomenon are not known. One hypothesis to explain the observed ranking proposes that the T allele
lowers the threshold for immune activation, consequently inducing a refractory state by raising the threshold and reducing the dynamic
range available for response to additional immunogenic stimuli from exogenous cytokines. Thymidine phosphorylase (TP) and cytidine
deaminase (CDA) are evolutionarily conserved enzymes of pyrimidine metabolism that are inducible directly and indirectly (respectively)
by pro-inflammatory cytokines. Increases in TP and CDA activity are therefore indicative of immune activation.
Hypothesis and Aim
If the IFN-L3 rs12971860 T allele is associated with increased immune activation, it may increase the activities of TP and CDA, which
are easily measurable in peripheral blood.
Methods
A simple HPLC-based assay for whole blood TP and CDA activity was devised and used to measure enzyme activities in >200 blood
samples obtained from individuals whose IFNL3 genotype was determined by a PCR-based method.
Results and Conclusions
Remarkably, concentrations of thymidine and cytidine close to a million-fold greater than their normal (submicromolar) plasma
concentrations were required to saturate blood TP and CDA. A very large (~10-fold) range of both activities was found. Correlation with
IFNL3 genotyping results revealed a ranked difference in enzyme activities in the order TT>CT>CC, supporting the hypothesis that the
rs1291860 T allele is associated with increased pro-inflammatory activity.
225
Spontaneous mutation in TNF in a new model of rheumatoid arthritis
Philippe Bouillet1, Derek Lacey1
1. Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
We have discovered mice with a genetic mutation that predisposes to rheumatoid arthritis. The trait is autosomal dominant and fully
penetrant. The affected mice present with a severe symmetrical, erosive chronic poly-arthritis, similar to human rheumatoid arthritis
(RA). It mostly affects the peripheral joints (the ankles, metatarsal and inter-phalangeal joints) with less severe damage in the central
joints (knees and shoulders). Inter-vertebral joints, especially between T9 and T13, are also affected. The bone marrow shows unusual
B lymphoid aggregates, associated with osteolytic lesions, and bronchus associated lymphoid tissue is present in all affected mice.
Homozygozity dramatically increases the severity of the disease. The phenotype is independent of T and B cells, and still develops in
the absence of Myd88 or GM-CSF.
We found that the mutation is a spontaneous insertion of a transposon in the 3’UTR of the TNF gene, and causes a dramatic increase in
the expression of this cytokine. Accordingly, loss of TNFR1 completely prevents all aspects of the phenotype.
We are presently in the process of identifying new regulators of TNF expression.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 75
226
Alterations in inflammatory cytokine network in diseased conditions associated with cognitive
impairments
Anna Boyajyan1, Martin Petrek2, Roksana Zakharyan1, Lilit Hovhannisyan1, Gohar Mkrtchyan1, Elina Arakelova1, Hovsep
Ghazaryan
1. Institute of Molecular Biology NAS RA, Yerevan, Republic of Armenia
2. Palacky University , Olomouc, Czech Republic
Background. Numerous findings suggest implication of upregulated inflammation in defects of neuronal plasticity and synaptic
connectivity leading to cognitive impairments in schizophrenia, posttraumatic stress disorder (PTSD) and ischemic stroke (IS). However,
molecular pathomechanisms responsible for development of deleterious inflammatory reactions in these diseased conditions are only
beginning to be understood. Aim. This report provides a brief overview of our recent studies on functional state of the major
inflammatory mediators, proinflammatory and chemotactic cytokines and their receptors, in etiopathogenesis of schizophrenia, PTSD,
and IS. Methods. Experiments were performed on gene, transcription, and protein levels using genomic DNA, mRNA, and blood plasma
samples of diseased and healthy individuals (250 subjects in each group). Single specific primer-polymerase chain reaction (SSPPCR), real-time PCR and enzyme-linked immunosorbent assay were applied. Data were analyzed using a wide range of methods
including parametric and nonparametric statistical tests, multiple comparisons corrections, and correlation analysis. Results. The results
obtained indicated association of polymorphic variations in genes encoding a number of proinflammatory and chemotactic cytokines and
their receptors with schizophrenia, PTSD and IS. Different types of associations were found for each disorder. In addition diseasespecific changes in gene expression levels as well as in plasma concentration for many proinflammatory and chemotactic cytokines and
their receptors were detected. Some of these changes correlate with frequency and intensity of clinical signs, symptoms, and severity of
diseased conditions as well as other clinically significant characteristics of patients such as age of the disease onset, etc. Also,
relationships between genotypes and protein expression levels were found for many cytokines. Conclusions. Schizophrenia, PTSD, and
IS are accompanied with changes in functional activities of proinflammatory and chemotactic cytokines and their receptors. Each
disease is characterized by specific combination of single nucleotide variations, changes in mRNA and protein expression patterns of
the inflammatory cytokine network components.
227
F-actin is required for negative regulation of NLRP3 inflammasome activity by Flightless-I and
LRRFIP2
Karim J Brandt1, Céline Fickentscher1, Philippe de Moerloose1, 2, Danielle Burger3
1. University of Geneva, Geneva, Switzerland
2. Angiology and Hemostasis, University Hospital of Geneva, Geneva, Switzerland
3. Immunology and Allergology, University Hospital of Geneva, Geneva, Switzerland
The NLRP3 inflammasome interacts with the adaptor ASC to activate caspase-1 for the maturation of pro-IL-1β into active IL-1β. The
formation and activity of NLRP3 inflammasome is regulated through mechanisms not fully elucidated. It was recently demonstrated that
leucine-rich repeat Flightless-I-interaction protein 2 (LRRFIP2) and Flightless-I (FliI) negatively regulate NLRP3 inflammasome activity.
It was also showed that FliI is a pseudosubstrate and inhibitor of caspase-1. Since FliI is an actin-remodeling protein, we addressed the
question of a role of actin in the regulation NLRP3 inflammasome in human macrophages (THP-1). The depolymerization of filamentous
actin (F-actin) in globular actin (G-actin) significantly increased the production of mature IL-1β while the stabilization of F-actin
decreased IL-1β production. These results suggest that activation of NLRP3 inflammasome depends on the actin polymerization state
but not on the active polymerization process. The silencing of LRRFIP2 and FliI by lentiviral-based shRNA transduction showed that
LRRFIP2 and FliI were required for co-localization of NLRP3 on F-actin in response to ATP or Nigericin. Thus, FliI-mediated caspase-1
inhibition involves the localization of NLRP3 inflammasome on F-actin leading to the diminution of caspase-1 activation and in turn to
decreased IL-1β production. The present data demonstrate that in addition to directly inhibiting caspase-1, FliI together with LRRFIP2
allow the location of the NLRP3 inflammasome on F-actin, further inhibiting caspase-1 activation and IL-1β production. Our results
unveil a new function of actin and a dual function of FliI in the regulation of NLRP3 inflammasome activity strengthening the importance
of cytoskeleton in the regulation of inflammation.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 76
228
Super-resolution microscopy reveals important roles for microtubule bundling by rabies virus P3
protein in immune evasion and disease
Aaron M Brice1, Donna R Whelan2, Kim Lieu3, Naoto Ito4, Linda Wiltzer3, Camden Y Lo5, Danielle Blondel6, Toby DM Bell2, David
A Jans3, Gregory W Moseley1
1. Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia
2. School of Chemistry, Monash University, Melbourne, Victoria, Australia
3. Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
4. Faculty of Applied Biological Sciences and the United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
5. Monash Micro Imaging, Melbourne, Victoria, Australia
6. Unité de Virologie Moleculaire et Structurale, CNRS, Gif sur Yvette Cedex, France
Rabies virus (RABV) is the causative agent of rabies, a severe neurological disease with a c.100% case-fatality rate that results in
>55,000 human deaths per year. The high lethality of RABV is thought to be dependent on its ability to inhibit host interferon (IFN)mediated antiviral immune signalling via the association of its IFN-antagonist P3 protein and STAT immune signalling proteins. In
vitro studies have indicated that P3 association with the microtubule (MT) cytoskeleton is important to STAT signalling inhibition but the
molecular details of the P3-STAT-MT interaction and its roles in disease in vivo are currently unresolved.
Using a unique RABV pathogenicity model, viral reverse genetics, immune signalling assays and a novel, quantitative in vivo MT tracing
assay using live 3D confocal laser scanning microscopy (CLSM), we identified a specific mutation (N226-H) that can impair the MTassociation, as detected by CLSM, and IFN-antagonistic function of P3. Introduction of the N226-H mutation into an invariably lethal
strain of RABV strongly impaired disease progression in vivo in mice. Importantly, analysis utilising super-resolution dSTORM
(directSTochastic Optical Reconstruction Microscopy) revealed that P3 induces significant MT bundling, and that N 226-H is defective in
this respect. However, N226-H does not affect the physical interaction of P3 with MT fractions or tubulin, indicating that the mutation
specifically impacts P3’s capacity to modify MT structure. This indicates a vital link between MT bundling and inhibition of immune
signalling by P3 and this mechanism is crucial to RABV pathogenicity, a novel discovery among viruses.
This data indicates that RABV and potentially other pathogenic viruses exploit/modulate dynamic MT functions to manipulate host cell
biology. This study has clear implications for our understanding of viral disease mechanisms and their potential targeting for
vaccine/therapeutic development and highlights the power of super-resolution microscopic approaches to probe molecular events at the
host-pathogen interface. Ongoing research aims to characterise in detail the molecular events at the MT interface of RABV, Ebola and
HIV-1.
229
The Innate Immune Sensor LGP2 Activates Antiviral Signaling by Regulating MDA5-RNA Interaction
and Filament Assembly
Annie M Bruns1, Robert A Lamb1, Curt M Horvath1, George P Leser1
1. Molecular Biosciences, Northwestern University, Evanston, IL, United States
Cytoplasmic pattern recognition receptors detect non-self RNAs during virus infections and initiate antiviral signaling. One receptor,
MDA5, possesses essential signaling domains, but weak RNA binding. A second receptor, LGP2, rapidly detects diverse dsRNA
species, but lacks signaling domains. Accumulating evidence suggests LGP2 and MDA5 work together to detect viral RNA and
generate a complete antiviral response, but the basis for their cooperation has been elusive. Experiments presented here address this
gap in antiviral signaling, revealing that LGP2 assists MDA5-RNA interactions leading to enhanced MDA5-mediated antiviral signaling.
LGP2 increases the initial rate of MDA5-RNA interaction and regulates MDA5 filament assembly, resulting in the formation of more
numerous, shorter MDA5 filaments that are shown to generate equivalent or greater signaling activity in vivo than the longer filaments
containing only MDA5. These findings provide a mechanism for LGP2 coactivation of MDA5 and a biological context for MDA5-RNA
filaments in antiviral responses.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 77
230
An elucidation of the regulation of the Interferon pathway using
the Interferome and Enrich bioinformatics resources.
Sam Forster1, Ross Chapman1, Helen Cumming1, Alex Finkel1, Jamie Gearing1, Kevin Luu1, David Hughes1, Paul Hertzog1
1. Centre for Innate Immunity & Infectious Diseases, MIMR-PHI, Monash University, Clayton, Victoria, Australia
Aims
Interferons play a vital role in mammalian responses to bacterial and viral infections and to tumor development. The Interferon response
following disease challenge is highly modulated. The host response must be sufficient to protect the host, but excessive responses can
cause toxicity and even death. Furthermore, the Interferon response follows a highly structured temporal response, with stimulation
activating an organised sequence of transcriptomic events. Here we present two bioinformatics tools that can be used to analyse the
transcriptional regulation of the Interferon response: 1) the Interferome v2.0 database
(http://interferome.its.monash.edu.au/interferome/home.jspx), which identifies genes regulated by Interferons from mouse and human
systems, and 2) the Enrich custom software, which is powerful and intuitive tool for identifying transcription factors involved in regulating
co-expressed gene sets and thus elucidating signaling pathways.
Methods
The Interferome database was queried for genes that were up-regulated in murine cells over discrete time periods following stimulation
by type I Interferons. Transcription factors that were significantly enriched (p<0.05) among each gene set were identified using
the Enrich custom bioinformatics tool.
Results
Interrogation of the Interferome database identified hundreds of genes that are up-regulated following stimulation by type I Interferon at
different time points. Following analysis of the associated promoters using Enrich, we have been able to identify clear patterns in the
transcription factors associated with temporally co-expressed genes. Many of the transcription factors identified are already known to be
involved in the Interferon pathway, such as STATs and IRFs. However, the analysis also revealed the involvement of further additional
transcription factors which might provide new insights into Interferon signalling.
Conclusion
By integrating the Interferome and Enrich bioinformatics resources, it has been possible to explore the transcription factors that regulate
different phases of the interferon response. The analysis has identified new regulatory elements for experimental validation.
231
Interleukin-37 – An Anti-inflammatory Cytokine for Necrotizing Enterocolitis?
Steven X Cho1, 2, Ina Rudloff2, Philip J Berger2, Wei Cheng3, 4, 5, 1, Marcel Nold2, Claudia Nold2
1. Monash University, Melbourne
2. The Ritchie Centre, MIMR-PHI Institute of Medical Research, Melbourne
3. Department of Paediatric Surgery, Monash Children’s, Southern Health. Department of Paediatrics, Department of Surgery,
Southern Medical School, Faculty of Medicine, Nursing of Health Sciences, Monash University, Melbourne, VIC, Australia
4. Department of Surgery, Beijing United Family Hospital, Beijing
5. Capital Institute of Pediatrics, Beijing
Introduction: Necrotizing enterocolitis (NEC) is sometimes dubbed the spectre of neonatal intensive care units (NICUs), as its onset is
insidiously non-specific and once NEC becomes manifest, the damage inflicted on the premature baby’s intestine and other organs is
often disastrous. It remains unknown which participants of the immune system initiate the cascade of events that lead to NEC;
consequently, no specific therapy exists. We hypothesize that an immature and inappropriate inflammatory response towards bacterial
colonization and/or food components plays a pivotal role in the initiation and perpetuation of NEC. Hence, we investigated the
therapeutic potential of the anti-inflammatory cytokine interleukin-37 (IL-37) in a murine model of NEC.
Methods: Newborn C57BL6J mice transgenic for IL-37 (IL-37tg) and wild-type (WT) controls (n=8 for both) were collected and
separated from dams at birth. Mice were formula-fed every 3hrs using the Yajima style Hoshiba nipple and subjected to asphyxia (100%
nitrogen gas for 45s) followed by cold stress (4°C, 5mins) twice daily to induce NEC. At the experimental endpoint (72hrs), all pups were
culled and intestinal samples collected. Littermates were dam-fed and kept as controls (n=16).
Results: On a 0-3 scale (no to severe pathology), clinical scores were 1.8 (WT) vs 1.2 (IL-37tg) for ileus, 0.9 vs 0.1 for hematochezia
and 1.5 vs 0.6 for diarrhea. Tissue histological injury scores assessing epithelial vacuolation and mucosal disintegration were 1.8 (WT)
vs 0.8 (IL-37tg) in the duodenum, 1.4 vs 0.6 in the jejunum and 1.4 vs 0.8 in the ileum. This marked reduction of NEC severity in IL-37tg
mice was associated with a decrease in IL-1β.
Conclusion: Our results demonstrate that IL-37 protects newborn mice against NEC, indicating that excessive inflammation contributes
to disease progression. Blocking this inappropriate inflammatory response may represent a promising approach to improve the outlook
of babies suffering from this devastating disease.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 78
232
Undefined region of ectodomain plays a role in determining the activity of TLR8
Tsung-Hsien Chuang1, Chao-Yang Lai1, Yi-Ling Liu1, Da-Wei Yeh1, Chih-Hao Lu1
1. National Heatlh Research Institutes, Miaoli, Taiwan
Toll-like receptors play important roles in detecting pathogen-associated molecular patterns for initiation of immune responses against
microbial infections. Toll-like receptor 8 (TLR8) belongs to a subfamily comprising TLR7, TLR8 and TLR9, and recognizes viral singlestranded RNA and small molecular weight agonists to activate anti-viral immune responses. TLR8s from different species have distinct
ligand recognitions. TLR8s from several non-rodent species including cat, horse, sheep, and bovine are activated by small molecular
TLR8 agonists, whereas TLR8s from mouse and rat, two rodent species, are activated only in the presence of PolyT-ODN. The lack of a
five-amino-acid motif in the undefined region following the leucine-rich repeat 14 of the mouse and rat TLR8 ectodomains has been
suggested to be a reason for the weak activities of these two rodent TLR8s. To further investigate the activation of TLR8, in this study
we cloned rabbit TLR8 (rabTLR8) cDNA. The rabTLR8 has a longer undefined region in its ectodomain than the TLR8 from other
species. In cell-based assay, this rabTLR8 is activated by TLR8 ligand in the absence of PolyT-ODN. Nevertheless, upon stimulation
the rabTLR8 had a lower activation compared to the activation of TLR8 from other species, except the mouse and rat TLR8s. Using
different deletion and human-rabbit chimeric TLR8 expressing constructs, we showed that the extra peptide in the undefined region
played a role in determining the activity of rabTLR8. These results suggest that the undefined region plays a role in determining the
activity of rabTLR8.
233
Proatherogenic Condition Promotes Autoimmune Th17 Cell Responses.
Yeonseok Chung1, Hoyong Lim1
1. Seoul National University, Seoul, SEOUL, South Korea
Patients with systemic autoimmune diseases show increased incidence of atherosclerosis. However, the contribution of proatherogenic
factors to autoimmunity remains unclear. We found that atherogenic mice (herein referred to as LDb mice) exhibited increased serum
interleukin-17, which was associated with increased numbers of T helper 17 (Th17) cells in secondary lymphoid organs. The
environment within LDb mice was substantially favorable for Th17 cell polarization of autoreactive T cells during homeostatic
proliferation, which was considerably inhibited by antibodies directed against oxidized low-density lipoprotein (oxLDL). Moreover, the
uptake of oxLDL induced dendritic-cell-mediated Th17 cell polarization by triggering IL-6 production in a process dependent on TLR4,
CD36, and MyD88. Furthermore, self-reactive CD4+ T cells that expanded in the presence of oxLDL induced more profound
experimental autoimmune encephalomyelitis. These findings demonstrate that proatherogenic factors promote the polarization and
inflammatory function of autoimmune Th17 cells, which could be critical for the pathogenesis of atherosclerosis and other related
autoimmune diseases.
234
West Nile virus sfRNA prevents incorporation of anti-viral molecules into exosomes to facilitate virus
spread
Brian D Clarke1, Jeff Wilusz2, Alexander A Khromykh1
1. Australian Infectious Disease Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland,
Brisbane, Queensland, Australia
2. Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, CO, USA
Exosomes and microvesicles are small 40 - 100 nm extracellular vesicles, which act as carriers for intercellular signalling. In particular,
these vesicles are packed with small RNAs, such as microRNAs, as well as proteins, and mRNAs. In this study, we show that
microvesicles derived from A549 cells stimulated with interferon (IFN) are potent inhibitors of West Nile virus (WNV) replication in cells
treated with these extracellular vesicles. siRNA knockdown of cellular RNases Xrn1, Dicer, Drosha, and RNaseL in IFN-stimulated cells
reduced the anti-WNV activity of extracellular vesicles. Anti-WNV activity was also observed for vesicles derived from cells infected with
WNV mutant IRAΔCS3 deficient in generation of subgeneric flavivirus RNA (sfRNA) but not for vesicles derived from wild type WNVinfected cells. We have previously shown that WNV sfRNA inhibits cellular RNA decay pathway by inactivating Xrn1 and RNAi pathway
by inhibiting Dicer. Therefore, we propose that WNV sfRNA inhibits generation of anti-viral RNA decay intermediates and/or miRNAs
and their subsequent incorporation into microvesicles and exosomes in order to facilitate virus spread. To test the hypothesis we are
undertaking RNAseq to identify exosomal RNAs from WT and IRAΔCS3 infected cells and will present the data at the meeting.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 79
235
Combined siRNA and TLR-activating therapy for the improved treatment of viral infections.
Daniel T.W. Clarke1, Jana McCaskill2, Glenn Marsh3, Paul Monaghan3, Linfa Wang3, Timothy Doran3, Nigel McMillan1
1. Griffith University, Gold Coast, QLD, Australia
2. Diamantina Institute, Brisbane, QLD, Australia
3. CSIRO Australian Animal Health Laboratory, Geelong, VIC, Australia
RNA interference is one of the fastest developing fields in biological science and has been demonstrated as a promising therapy for a
wide range of viral infections. This highly specific suppression can be achieved by the introduction of short interfering (si) RNAs into
mammalian systems, resulting in the efficient reduction of viral load in vitro. However, this activity can have the unfortunate
consequence of non-specifically activating several innate immune sensors including toll-like receptor (TLR) 7/8 and stimulate the
expression of interferon, which can mask the true specific effects of the siRNA. While often not desired, the localised activation of TLR
pathways in combination with specific anti-viral activity of the siRNA may have a beneficial role in providing a more potent control of viral
infection, especially in reducing transmission between hosts. Here we show that specific TLR agonists are able to induce cytokines of
the innate immune response and improve the anti-viral response when used in tandem with siRNAs targeting specific viral sequences in
a Semliki Forest Virus model. Furthermore, individual pre-treatment of HeLa cells with poly(I:C) or viral nucleocapsid siRNA before
Hendra virus (HeV) infection resulted in a 1.5 log (>95%) reduction while the combination resulted in a 2.5 log (>99.9%) reduction in
titre, indicating a trend for the two pre-treatments to act simultaneously to cause additional suppression of HeV. This foundation will
allow further investigation into in vivo systems and once matched with efficient delivery mechanisms may result in better viral control
and a new treatment modality for acute viral infections.
236
Cleavage and Secretion of Interleukin-1b in the Absence of Cell Death.
Stephanie Conos1, James Vince1, David Vaux1, Lisa Lindqvist1
1. Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
Interleukin-1b (IL-1b) is an important signalling molecule in inflammation and auto-immunity. Specific host, environmental, and pathogen
derived danger molecules activate inflammasome protein complexes. These in turn activate caspase-1, which processes IL-1b into its
mature biologically active form. Active caspase-1 also induces lytic cell death known as pyroptosis. Whether pyroptosis and IL-1b
maturation and secretion are separable events remains unclear.
To study caspase-1 function in the absence of inflammasome signalling we created a lentiviral flag-tagged caspase-1-bacterial DNAgyrase-GFP fusion construct (FC1GG). Expression of the caspase-1 fusion protein is induced through addition of doxycycline. The
dimeric antibiotic coumermycin causes the gyrase domains to dimerise and caspase-1 to auto-activate.
Mouse embryonic fibroblast (MEF) cell lines engineered to constitutively express inactive precursor IL-1b were stably infected with the
inducible FC1GG construct. We observed that these cells, which do not express other inflammasome components, secreted IL-1b upon
caspase-1 induction and dimerisation. Notably, caspase-1 dependent IL-1b cleavage and secretion occurred in the absence of caspase1-mediated cell death.
This novel system allows the study of caspase-1-mediated IL-1b secretion without the activation of upstream signalling pathways, or
caspase-1 mediated pyroptosis. Consequently, we are currently using it to identify the cellular machinery required for IL-1b secretion via
mass-spectrometry based approaches.
237
MCC950 is a potent and specific inhibitor of the NLRP3 inflammasome and a novel therapeutic for
NLRP3 driven diseases
Rebecca C Coll1, Avril AB Robertson2, Jae J Chae3, Sarah C Higgins1, Lara S Dungan1, Brian G Monks4, Eicke Latz4, Kingston G
Mills1, Seth L Masters5, Matthew A Cooper2, Luke AJ O'Neill1
1. School of Biochemistry &Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Dublin 2, Ireland
2. University of Queensland, St Lucia, QLD, Australia
3. Inflammatory Disease Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of
Health, Bethesda, Maryland, United States of America
4. Institute of Innate Immunity, University Hospitals, Biomedical Centre, University of Bonn Bonn, Germany
5. The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Victoria, Australia
NLRP3 is a critical component of the inflammatory process and its aberrant activation is pathogenic in inherited disorders such as the
cryopyrin associated periodic syndromes and complex diseases such as multiple sclerosis, type II diabetes and atherosclerosis. We
demonstrate that a small molecule, MCC950, specifically blocks NLRP3 activation in response to numerous stimuli. MCC950 potently
inhibits ASC oligomerisation, caspase-1 activation and IL-1ß secretion in response to NLRP3 but not AIM2, NLRC4 or NLRP1
inflammasome activation. MCC950 can block IL-1ß in vivo and attenuates the course of the NLRP3 dependent, IL-1ß driven disease
model of multiple sclerosis, experimental autoimmune encephalomyelitis. Furthermore, MCC950 treatment rescues neonatal lethality in
a murine model of cryopyrin associated periodic syndromes and is active in ex vivo samples from patients with Muckle-Wells syndrome.
MCC950 is thus a novel potential therapeutic for NLRP3 associated syndromes and a useful tool for the further study of NLRP3 in
human health and disease.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 80
238
Interferon-α-induced IL-6 improves immune and clinical benefits of regulatory T cell depletion as
ovarian cancer immunotherapy
Suzanne R. Thibodeaux1, Srilaksmi Pandeswara2, Shawna Wall2, Vincent Hurez2, Lishi Sun2, Benjamin J. Daniel2, Ai-Jie Liu2,
Leslie Wood2, Weiping Zou3, Tyler J. Curiel2
1. University of Pennsylvania, Philadelphia, PA, USA
2. UT Health Science Center, San Antonio, TEXAS, United States
3. University of Michigan, Ann Arbor, MI, USA
Tregs blunt anti-tumor immunity. Their depletion is effective treatment in mouse cancer models. Treg depletion in human trials is only
partially effective, however. In our phase II ovarian cancer trial, denileukin diftitox (DT) depleted Tregs significantly in blood and the
tumor microenvironment but without significant clinical efficacy. Interferon-α alone does not treat ovarian cancer effectively, but we now
show that it significantly boosts immune and clinical effects of DT-mediated Treg depletion in human ovarian cancer. In the ID8 mouse
ovarian cancer model, DT improved anti-tumor immunity and survival modestly. Interferon-α alone did not affect Treg function or
numbers, but improved CD8+ T cell anti-tumor immunity. Adding interferon-α to DT significantly increased mouse survival versus either
agent alone. Using type I IFNR-/- mice that are unable to mediate interferon-α signals, we showed that interferon-α boosted CD8+ T cell
function independent of CD4+ T cell help through direct action on CD8+ T cells. In combination with DT, interferon-α reduced Treg
function (without further reducing their numbers) through indirect effects on dendritic cells in vivo. In vitro we determined that interferonα reduced Treg suppression and increased IL-6 in Treg suppression assays only when conventional dendritic cells were present. AntiIL-6 antibody reversed the effect, and recombinant IL-6 recapitulated the effect in the absence of dendritic cells. When three ovarian
cancer patients failed DT alone in the phase II trial, two experienced immunologic and clinical benefit by adding weekly pegylated
interferon-α2a at 180 μg subcutaneously, with manageable toxicities. There was a trend for pegylated interferon-α2a to reduce Treg
function and increase serum IL-6 when combined with DT, versus effects of DT alone in these 3 patients. IL-6 is generally detrimental to
anti-tumor immunity and promotes carcinogenesis, but in specific instances it could be beneficial. These studies demonstrate novel
immune and clinical interferon-α anti-cancer benefits that augment Treg depletion using approved agents for rapid clinical translation.
239
Pathogenic potential of interferon αβ in acute influenza infection
Sophia Davidson1, Stefania Crotta1, Teresa M McCabe1, Andreas Wack1
1. National Institute for Medical Research, London, United Kingdom
Severe influenza virus-induced disease is characterised by acute lung injury, associated with a vigorous inflammatory response.
However, disease severity varies between individuals and this cannot be entirely explained by differences in influenza strain virulence,
or an individual’s pre-existing adaptive immunity or health status. Host specific genetic factors must therefore contribute to susceptibility.
Type I interferon (IFNαβ) is known to have antiviral function in-vitro, yet its role in influenza infection in-vivo is less clear. Aims: To
identify host specific determinants of susceptibility to influenza-induced disease. Methods: We assessed susceptibility of inbred mouse
strains to influenza-induced disease where viral load was comparable during early infection. Influenza-induced gene expression,
cytokine production and cellular recruitment, activation and death in the pulmonary environment were compared between susceptible
and resistant mouse strains. Results: Influenza infected 129 and DBA strains showed dramatically increased morbidity, mortality and
lung damage, yet higher levels of pulmonary IFNαβ, compared to the more resistant C57BL/6 or BALB/C mice. Ablation of IFNαβR
signalling in 129 mice markedly reduced mortality, lung damage, proinflammatory cytokines and lung-infiltrating inflammatory cells such
as inflammatory monocytes (IMs). Furthermore, IFNα treatment of influenza-infected C57BL/6 mice increased morbidity. Plasmacytoid
dendritic cells (pDCs), which are potent IFN producers, were recruited in higher numbers to lungs of infected 129 mice. pDC derived
IFNαβ was demonstrated to be upstream of the robust proinflammatory response. Significantly, IFNαβ signalling induced the
upregulation of Tumour necrosis factor-Related Apoptosis-Inducing Ligand (TRAIL) on IMs and its receptor: Death Receptor 5 (DR5) on
lung epithelia. TRAIL-DR5 interaction is required for epithelial apoptosis, lung damage and mortality of 129 mice. Conclusion: Hostintrinsic differences can determine the severity of influenza-induced disease. pDC frequency and responsiveness to IFNαβ signalling is
a host-specific determinant with protective or pathogenic potential. This study has important implications for the prediction and treatment
of severe influenza-induced disease.
240
Development and characterization of a cell-penetrant IRF5 inhibitor
Saurav De2, 1, De Feng2, Betsy Barnes2
1. Rutgers Graduate School of Biomedical Sciences, Newark, , New Jersey, USA
2. Rutgers Biomedical and Health Sciences University, Newark, NJ, United States
The transcription factor interferon regulatory factor 5 (IRF5) has previously been implicated in the onset of the autoimmune disorder
systemic lupus erythrematosus (SLE). Elevated levels of inflammatory cytokines are a common characteristic of SLE, and are believed
to contribute to both autoantibody production and wide spread inflammation. Upon activation, cytoplasmic IRF5 translocates to the
nucleus to initiate pro-inflammatory gene transcription. To achieve nuclear translocation, IRF5 relies on two nuclear localization signals
located in the N’ and C’ termini of the protein. To investigate the therapeutic potential of IRF5 inhibition, we have developed two unique
cell-penetrating peptides. Upon treatment with these inhibitors, we show IRF5 is excluded from the nucleus, while IRF7 and NFkB
nuclear translocation were unaffected following activation. We have investigated the impact of IRF5 inhibition in a variety of cell lines as
well as primary peripheral blood mononuclear cells. The inhibitors show no impact on cell cycle, viability, or IRF5 protein levels. As
IRF5 has previously been linked to expression of IgG subtypes in mice, we utilized the IRF5 inhibitors to examine the impact of IRF5
inhibition in the Ramos B cell line. Interestingly, we found no impact of the inhibitors on surface IgG expression. In THP1 cells,
however, a marked reduction in inflammatory cytokine expression was seen following stimulation with LPS and IFNg. These data
highlight the potential of targeting IRF5 in order to reduce the inflammatory signature characteristic of SLE patients.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 81
241
Characterizing the role of IRF5 in human B cell development and function
Saurav De1, 2, Di Feng1, Peicheng Du3, Robert Donnelly4, 5, Betsy Barnes1
1. Rutgers Biomedical and Health Sciences University, Newark, NJ, United States
2. Rutgers Graduate School of Biomedical Sciences, Newark, , New Jersey, USA
3. High Performance and Research Computing Group, Office of Information Technology, Rutgers University, Newark, New Jersey,
USA
4. Department of Pathology and Laboratory Medicine and NJMS Molecular Resource Facility, Rutgers Biomedical and Health
Sciences, Newark, New Jersey, USA
5. Department of Pathology and Laboratory Medicine and NJMS Molecular Resource Facility, Rutgers Biomedical and Health
Sciences, Newark, New Jersey, USA
The transition of naïve B cells to effector B cells is dependent on a large transcription factor network, which mediates both effector B cell
differentiation and function. The full repertoire of transcription factors involved in this process is not known, yet dysregulation of this
transcription factor network can result in altered B cell function and autoimmunity. Work from our lab, as well as others, has suggested
that the transcription factor, interferon regulatory factor 5 (IRF5), is involved in the development of effector B cells. Irf5-/-mice have
previously been reported to have reduced plasma B cells, as well as reduced serum IgG subtypes. It remains unclear, however, what
role IRF5 may play in human B cell development and function. We find significant levels of IRF5 in B cells translocate to the nucleus
following stimulation with anti-IgM antibody and CpG. In order to characterize the role of IRF5 in human B cells, we have performed
IRF5 ChIP-Seq in both primary naïve B cells and Ramos B cells either mock or anti-IgM and CpG treated. Genes associated with
plasma B cell development were significantly enriched following activation, suggesting IRF5 plays a critical role in the differentiation of
plasma B cells. To further characterize the role of IRF5 in primary human B cells, we have been able to successfully perform siRNAmediated knockdown of IRF5. Knockdown of IRF5 did not show significant impact on cell viability, however, reduced inflammatory
cytokine expression was seen. These data highlight the multi-functional role of IRF5 in regulating both human B cell differentiation and
function.
242
Role of an IFNAR1-IFNβ mediated signalling axis in immune cells
Nicole A De Weerd1, Julian Vivian2, Thao Nguyen1, Niamh Mangan1, Jodee Gould1, Susie Braniff1, Leyla Zaker-Tabrizi1, KaYee
Fung1, Sam Forster1, Travis Beddoe2, Hugh Reid2, Jamie Rossjohn2, Paul Hertzog1
1. MIMR-PHI Institute, Clayton, Victoria, Australia
2. Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
Type I interferons (IFNs) are an important family of cytokines which enable the immune system to fight viral infections and cancer, and
modulate the immune response. Type I IFNs are unique amongst cytokines since multiple ligands can signal through the same
heterodimeric receptor composed of low (IFNAR1) and high (IFNAR2) affinity components. Despite sharing a receptor, discernible
differences result from receptor engagement by different IFN subtypes. We recently demonstrated a molecular basis for the unique
functional characteristics exhibited by IFNβ; it can bind and signal via IFNAR1 in the absence of IFNAR2, inducing a novel signaling axis
that contributes to lethality in a mouse model of sepsis. Having determined the crystal structure of IFNβ bound to full length extracellular
domain (ECD) of IFNAR1, we are validating the interface to identify key residues mediating this interaction, and further characterising
the signaling pathway. The novel signaling axis we identified by microarray analysis of induced ISGs after in vivo IFNβ treatment of
IFNAR2-/-mice includes genes encoding proteins with known roles in sepsis. We now demonstrate that this treatment also induces
phosphorylation of AKT in cells of the peritoneal cavity. Furthermore, we observed reduced levels of B220+CD11c- leukocytes in the
peritoneal cavity and increased presentation of TREM1 on the surface of Gr1+ peritoneal cells in an IFNAR1-IFNβ dependent manner.
That TREM1 is known to amplify the lethal response associated with sepsis gives some mechanistic insight into how the IFNAR1-IFNβ
signaling axis may be contributing to lethality in this disease. Our results further demonstrate the importance of IFNβ and IFNAR1 in the
transmission of signals and in influencing cell numbers and increased surface expression of inflammatory mediators. Understanding
how to abrogate IFNβ binding to IFNAR1, and the down stream effects of the IFNAR1-IFNβ signaling axis may aid in development of
targeted therapeutics in IFNβ-mediated inflammation.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 82
243
Targeting placental leukemia inhibitory factor in vivo with a unique inhibitor as a novel treatment
strategy for ectopic pregnancy.
Amy Winship1, Tara Krishnan1, Ellen Menkhorst1, Andrew Horne2, Jeremy Brown2, Stephen Tong3, 4, Jianguo Zhang5, Nick
Nicola5, Evdokia Dimitriadis1
1. MIMR-PHI Institute, Clayton, VIC, Australia
2. The University of Edinburgh, Edinburgh, Scotland, UK
3. The University of Melbourne, Melbourne, VIC, Australia
4. The University of Melbourne, Melbourne, VIC, Australia
5. Walter and Eliza Hall Institute, Melbourne, VIC, Australia
Ectopic pregnancy is unique to humans and a leading cause of maternal morbidity and mortality. The etiology remains unknown
however factors regulating embryo implantation likely contribute. Leukemia inhibitory factor (LIF) has roles in extravillous trophoblast
adhesion and invasion during normal placental development and is also present in ectopic implantation sites. We hypothesised that LIF
facilitates blastocyst adhesion/invasion in the Fallopian tube, contributing to ectopic pregnancy. LIF blockade could serve as a potential
treatment strategy.
LIF receptor (R) was immunolocalised in tubal ectopic pregnancy implantation sites (N=5). An oviduct cell line (OE-E6/E7) and a
trophoblast cell line (HTR-8/SVneo; spheroid culture) were used to model blastocyst attachment to the Fallopian tube. LIF signaling
pathways in OE-E6/E7 were examined by Western blot. The effect of LIF and LIF inhibition (using PEGylated LIF antagonist; PEGLA)
on first-trimester placental outgrowth was determined. To demonstrate that LIF blockade could reverse trophoblast invasion in
vivo,pregnant C57BL/6J mice were IP administered with PEGLA (500µg/application PEGLA or PEG control) at 1000h and 1600h on
days (D)8-10 or 10-13 of pregnancy (D0: day of plug). Implantation sites collected at D10 or 13 were stained with cytokeratin
(trophoblast marker) isolectin-B4 and α-SMA (vascular markers).
LIFR localised to villous and extravillous trophoblast and Fallopian tube epithelium in ectopic pregnancy. LIF activated STAT3 but not
the ERK pathway in OE-E6/E7 cells. In vitro, PEGLA inhibited LIF stimulated HTR-8/SVneo spheroid adhesion to OE-E6/E7 cells and
explant outgrowth. LIF inhibition in pregnant mice impaired decidualization and placental spongiotrophoblast and labyrinth formation.
Our data suggests LIF facilitates the development of ectopic pregnancy by stimulating blastocyst adhesion and trophoblast outgrowth
from placental explants. In mice, LIF inhibition dramatically altered placental development and trophoblast structure. Ectopic pregnancy
is usually diagnosed after 6 weeks of pregnancy, therefore PEGLA may be useful as a treatment for ectopic pregnancy.
244
Induction of IL-6 expression through TLR4 by oxidised phospholipids involves the selective
induction of Arid5a but not Regnase-1 expression.
Praveen Dubey1, Kazuya Masuda1, Kishan Nyati1, Barry Ripley1, Tadamitsu Kishimoto1
1. Osaka University, Osaka, Japan
Pathology following influenza infection develops through the induction of host-derived oxidized phospholipids (OxPLs), which stimulate
TLR4-dependent inflammation and macrophage-IL6-dependent acute lung injury. Interestingly, through TLR4 stimulation in alveolar
macrophages, lipopolysaccharide (LPS) induces expression of the pro-inflammatory cytokines IL-6 and TNF-α, however OxPLs induce
only IL-6 expression.
The mechanistic basis for the selective induction of IL-6 by OxPLs (compared to LPS) is not known, but is clearly of importance to
understanding the pathogenesis of influenza-induced lung pathology.
Recently, we identified an LPS-TLR4-induced protein called Arid5a, which selectively stabilizes IL-6 (but not TNF-α) mRNA and
counteracts the function of the LPS-TLR4-induced RNAse Regnase-1 which degrades IL-6 mRNA. In the current study, using cultured
peritoneal macrophages, we have found that through TLR4 signaling, OxPLs induce Arid5a, but do not induce Regnase-1 expression.
We also observed that treatment with OxPLs inhibit the basal mRNA levels of TNF-α but not IL-6. Interestingly, in co-stimulation
experiments, OxPLs inhibited the expression levels of TNF-α (but not IL-6) mRNA following stimulation of peritoneal macrophages with
either poly I:C (TLR3 ligand) or CpG-a (TLR9 ligand).
Taken together, these observations indicate that OxPLs selectively induce IL-6 expression through TLR4 through two biological
properties (i) promoting IL-6 mRNA stability through induction of Arid5a but not Regnase-1 and (ii) by inhibiting TLR4-TNF-α mRNA
expression levels. The molecular mechanisms underlying these observations and their involvement in lung pathology, are currently
under investigation.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 83
245
IL-7R: a target in ALL and autoimmunity
Scott Durum1, W. Q. Li1, E. Senkevitch1, P. Zenatti2, D. Ribeiro3, L. Zuurbier4, M. C. Silva3, M. Paganin5, J. Tritapoe1, J. A. Hixon1,
A. B. Silveira2, B. A. Cardoso3, L. M. Sarmento3, N. Correia3, M. L. Toribio6, J. Kobarg7, M. Horstmann8, 9, R. Pieters4, S. R.
Brandalise2, 10, A. A. Ferrando5, J. P. Meijerink4, J. A. Yunes2, 11, J. T. Barata3
1. National Cancer Institute, Frederick, MD, United States
2. Centro Infantil Boldrini, Brazil
3. Universidade de Lisboa, Portugal
4. Erasmus Medical Center, The Netherlands
5. Columbia University, USA
6. Universidad Autónoma de Madrid, Spain
7. Centro Nacional de Pesquisa em Energia e Materiais, Brazil
8. German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Germany
9. University Medical Center Hamburg-Eppendorf, Germany
10. Universidade Estadual de Campinas, Brazil
11. Universidade Estadual de Campinas, Brazil
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy resulting from leukemic transformation of T-cell
progenitors in the thymus. It accounts for approximately 15% of ALL cases in childhood and 20-25% in adults and is a leading cause of
death in children. IL-7 and its receptor (IL-7R) play a critical role in normal T-cell development and homeostasis. Mutations in IL-7R
were identified in 9% of pediatric T-ALL patients. These mutations usually involved insertions of three amino acids including cysteine
and proline in the extracellular juxtamembrane region. WT or mutant forms of the human IL-7R (hIL-7R) from patients were retrovirally
transfected into an IL-7-dependent murine thymic cell line D1. Mutant hIL-7Rs induced ligand-independent activation of the Jak-Stat and
PI3K pathways, cell survival and proliferation. Constitutive signaling required homodimerization via cysteines in the inserted sequences
and downstream Jak1 activation, and was IL-7, gc and Jak3-independent. Mutant hIL-7R-expressing D1 cells formed subcutaneous
tumors in Rag1-/- mice, with substantial infiltration into various organs that are normally affected in advanced stages of T-ALL, such as
bone marrow, liver, lymph nodes and spleen. Janus kinase inhibitors effectively killed these cells in vitro and in vivo. The hotspot for
insertions lies in exon 6 in precisely the same region as a coding polymorphism regulating risk for MS and other autoimmune diseases,
and we observe that this polymorphism affects strength of signaling. Our findings indicate that IL-7R mutations drive T-ALL, whereas
polymorphisms that increase signaling promote autoimmunity, implicating IL-7R and Jak1 as therapeutic targets in these diseases.
246
Cytokine dysregulation in autoimmune disease pathogenesis
Julia Ellyard1
1. John Curtin School of Medical Research, Acton, 0200, Australia
There are more than 80 different autoimmune diseases, and collectively they affect 3-5% of the population. They are chronic, incurable,
and cause substantial morbidity and mortality. The fundamental lack of understanding of autoimmune disease pathogenesis means that
available treatments – most causing general immune suppression - are non-specific, often fail to treat the most serious disease
manifestations, and cause serious side-effects. Our research over the past decade first focused on “mouse-to-human” approaches to
understand autoimmunity. Random mouse mutagenesis led to our discovery of the Roquin family revealing a pathway in which
accumulation of Tfh cells caused by Roquin mutations leads to rogue positive B cell selection and autoantibody-mediated autoimmune
disease. Roquin and its paralogue Roquin2 act in T cells and myeloid cells to repress mRNAs posttranscritptionally. The key pathogenic
pathways leading to autoimmunity and inflammation when Roquin is mutated consist on excessive IFN-g production that promote Tfh
and GC B cell accumulation, and increased myeloid cell-derived TNF. More recently, we have undertaken a “human-mouse-human”
approach to focus our efforts on illuminating the mechanism of disease in each individual patient and elucidating the biological pathways
that connect human genetic variation to disease. This has led to identification of rare gene variants contributing strong effects to
autoimmune susceptibility. Amongst these is a case of childhood cerebral SLE manifesting in the first years of life with severe clinical
manifestations including stroke. A novel homozygous variant in TREX1 leading to a significant reduction in the protein’s exonuclease
activity causes increased type I interferon production, likely to be at the root of the autoimmune phenotype. This human-centered
approach is expected to help stratify what are notoriously heterogeneous diseases, and illuminate targeted therapies based on the
affected molecular pathways.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 84
248
Small tumor necrosis factor-α receptor (TNFR) biologics selectively inhibit the TNF-p38 signalling
axis and inflammation
Violet R Mukaro1, 2, Michelle E Gahan3, Bernadette Boog1, Alex Quach1, 2, Zia H Huang1, 2, Xiuhui Gao1, 2, Carol Haddad1, Suresh
Mahalingam4, Charles S Hii1, 2, Antonio Ferrante1, 2, 5, 6
1. SA Pathology at Women's and Children's Hospital, North Adelaide, SA, Australia
2. School of Paediatrics and Reproductive Medicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia,
Australia
3. Virus and Inflammation Research Group, Faculty of Applied Science, University of Canberra, Canberra, Australian Capital
Territory, Australia
4. Emerging Viruses and Inflammation Research Group, Institute of Glycomics, Griffith University, Gold Coast, Queensland, Austalia
5. Microbiology and Immunology, University of Adelaide, Adelaide, South Australia, Australia
6. School of Pharmacy and Medical Science, Univeristy of South Australia, Adelaide, South Australia, Australia
INTRODUCTION: Tumor necrosis factor (TNF) neutralising biologics, antibodies and soluble TNF receptors (TNFR), have
revolutionised treatment of diseases such as rheumatoid arthritis. However, targeting a cytokine with a key role in immunity to infection
and malignancy continues to raise serious concerns. Attempts to target the pro-inflammatory p38 MAP kinase, downstream of TNFR,
have ceased owing to poor efficacy and unacceptable toxicity. The aim of this research was to develop biologics that could selectively
inhibit the TNFR-p38 signalling axis and inflammation.
METHODS: Two TNF-derived peptides, TNF70-80 and TNF132-150 were investigated for their effects on neutrophil activation
(lucigenin-dependent chemiluminescence assay), tumour cell cytotoxicity (MTT assay) and activation of MAP kinases (western blot). A
pre-B cell line transfected with truncated TNFRI mutants was used to identify the region on TNFRI which binds TNF70-80. Peptides to
this region of TNFRI were made and investigated in mice for their ability to inhibit LPS-induced peritoneal inflammation, carrageenanand antigen-induce paw swelling/thickness and respiratory syncytial virus -induced lung inflammation, by assessing cell types and
number and cytokines present at the inflammatory sites.
RESULTS: We have previously demonstrated that, TNF70-80 and, TNF132-150 had distinct biological properties but which were
collectively representative of the parent TNF. Our data show that these differences reflect the types of signalling pathways they
stimulate via TNFRI. Of interest was that TNF70-80 selectively stimulated p38 activation. Using this peptide and truncated TNFR
mutants we identified the sequence in TNFRI which is coupled selectively to p38 activation. Peptides of this TNFRI sequence bind
TNF/TNF70-80 and selectively inhibited their ability to induced p38 activation, TNF-induced neutrophil stimulation and inflammation in
all four experimental models.
CONCLUSIONS: A TNFRI peptide which binds to TNF and selectively inhibits the TNFRI-p38 signalling axis and inflammation has been
developed as potentially a new approach to targeting TNF in inflammatory diseases.
249
Regulation of the Interferon Response by the long non-coding RNA lincRNA-EPS
Kate Fitzgerald1
1. Division of Infectious Diseases & Immunology, , University of Massachusetts Medical School, Worcester, MA, USA
Details not available at time of print...
250
Imidazoquinolines signal through TLR7 and IRF-5 for the production of type I and III interferon in
primary human plasmacytoid dendritic cells
Patricia Fitzgerald-Bocarsly1, Mahwish Natalia1, Jihong Dai1, Sukhwinder Singh1, Betsy Barnes1
1. Rutgers - New Jersey Medical School, Newark, NJ, United States
Human pDC recognize Influenza virus, HIV-1 and small molecules from the imidazoquinoline family through TLR7 and HSV and CpGA
through TLR9, resulting in production of type I and III IFN and pro-inflammatory cytokines. Imidazoquinolines have been used for antiviral therapy and as immune adjuvants and are often considered to be prototypical TLR7 agonists. However, there have been reports of
these molecules inhibiting virus-induced cytokine production. We investigated the mechanisms by which the imidazoquinoline, 3M003,
induces type I and III IFN production in pDCs while inhibiting virus-stimulated cytokine production. 3M003 induced Type I and III IFN and
TNF-ɑ much more rapidly than Flu or HSV, but at much lower levels and for shorter duration; actin polymerization and endosomal
acidification were required for this activation. The TLR7 inhibitor IRS661 effectively inhibited Flu and 3M003-induced IFN production.
3M003 inhibited Flu, HSV, HIV and CpGA induced IFN production in pDC when used together with these TLR7/9 agonists. This
inhibitory effect of 3M003 was observed when it was added any time in the first four hours of virus stimulation. Similar to Flu and HSV,
3M003 enhanced IRF7 serine 477/479 phosphorylation but, surprisingly, did not translocate IRF7 to the nucleus as shown by imaging
flow cytometry. Instead, 3M003 and the related molecule, R848, strongly and rapidly (within 30 minutes) induced IRF5 nuclear
translocation in pDC, while HSV induced strong IRF7 and to a similar extent, IRF5 translocation, but later. Co-culture of pDC with
3M003 and virus yielded the high IRF5/low IRF7 translocation phenotype. HSV-1- but not 3M003-induced IFN was inhibited by the PI3K
inhibitor, LY294-002. Taken together, these results indicate a novel signaling pathway in pDCs wherein 3M003 or 3M003 plus virus
preferentially activates IRF-5 in a PI3K-independent manner, while HSV activates both IRF-5 and -7, as well as PI3K, resulting in the
different levels and kinetics of IFN production.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 85
251
Comprehensive Integration of RNA and HITS-CLIP Sequencing for fine scale mapping of the Type 1
Interferon Response
Sam Forster1, Jodee Gould1, Michelle Tate1, Rebecca Piganis1, Ka yee Fung1, Alex Finkel1, Violeta Estrada2, Alexander Drew1,
Michael Gantier1, Nina Papavasiliou2, Kate Jeffrey3, Paul Hertzog1
1. Monash Institute of Medical Research, Clayton, VIC, Australia
2. Laboratory of Lymphocyte Biology, Rockefeller University, New York, USA
3. Harvard Medical School, Boston, USA
The innate immune response of an organism is the primary response to challenges including inflammatory stimuli, infectious agents or
the presence of cancer cells. The important protective role of IFN activation and signalling is a key component of this response. While
IFN signalling is essential for host survival, fundamental to vaccine responses, can be activated therapeutically and is often the target
for pathogen evasion strategies uncontrolled or aberrant responses can be toxic and result in disease. It is therefore critical to balance
signaling pathways to attain benefit while minimizing toxicity. While there is considerable knowledge of transcriptional regulation and
role of protein coding transcripts in the IFN response equivalent knowledge of non-coding responses and their regulatory mechanism
remains an active research area. In this context, an understanding of the role of small non-coding RNA, particularly miRNA, is rapidly
being gained with important insights in many areas of biology.
High throughput sequencing technologies have unlocked the ability to measure the complete transcriptional landscape of a cell
ultimately revolutionizing our ability to understand these complex systems. Combined with HITS-CLIP sequencing, a recently developed
experimental sequencing based approach capable of detecting microRNA regulatory interaction with target genes can provide important
regulatory insights into this system. We present the results of parallel IFNβ time course stimulation of small, ribosomal depleted total
RNA and HITS-CLIP sequencing complemented with miRNA expression and target network identification and experimental validation in
murine bone marrow derived macrophages. Analysis of this data has resulted in comprehensive network level understanding of the
complete transcriptional response to IFNβ including the identification of almost 100, IFNβ inducible putative novel miRNA and
associated characterization of potential targets. Novel computational algorithms and analysis approaches within our custom designed
RNA-eXpress framework have been developed and applied resulting in the generation of predictive target networks. Results of
experimental validation of these related miRNA pathways will also be presented.
This systems level interpretation of IFN mediated signaling has the potential to provide a basis for novel insights into both host-pathogen
interactions and host response critical in the efficient development of effectively targeted therapeutics.
252
Modulation of RIG-I signalling by OASL
Hans Henrik Gad1, Mikkes Søes Ibsen1, Line Lykke Andersen1, Saumendra Sarkar2, Rune Hartmann1
1. Aarhus University, Aarhus C, Denmark
2. University of Pittsburgh, Pittsburgh, USA
The innate immune system is the first line of defense against invading pathogens such as viruses. A fast and efficient innate immune
response depends upon an early recognition of the pathogen by so-called pattern-recognition receptors (PRRs). One of the most
important PRRs in terms of recognition of viral RNAs in the cytoplasm is the retinoic acid inducible gene I (RIG-I). Following viral
infection, RIG-I predominantly recognizes short double-stranded RNAs (dsRNA) containing triphosphate at their 5’ end and triggers a
distinct signal transduction pathway leading to induction of cytokines such as interferon (IFN) as well as other antiviral proteins.
Activation of RIG-I is regulated at several levels by cellular proteins in order to both prevent undue activation of the innate immune
response and to increase the efficiency of RIG-I-mediated signaling. For example, following recognition of viral RNA, RIG-I binds free
unanchored K63-linked polyubiquitin chains and this interaction with polyubiquitin is required for its activation. However, our collaborator
Saumendra N. Sarkar from the University of Pittsburgh Cancer Institute, USA, has recently discovered that another IFN-stimulated
protein known as the oligoadenylate synthetase-like protein (OASL) can act as a substitute for polyubiquitin in activation of RIG-I.
Through its two C-terminal ubiquitin-like repeats, OASL interacts with the RIG-I caspase recruitment domains (CARD) and thus mimics
the role of polyubiquitin. However, how the remaining N-terminal part of OASL contributes to the activation of RIG-I is not clear although
it was shown to interact with RIG-I as well. Here, we show that the N-terminal domain of OASL binds dsRNA and that this binding is
required in order for OASL to promote RIG-I activation. This suggests that OASL acts not only as a cofactor for RIG-I activation but also
as a PRR.
253
Master regulation of transcriptional responses by microRNAs
Michael P Gantier1
1. MIMR-PHI Institute of Medical Research, Clayton, VIC, Australia
microRNAs (miRNAs) are critical to most cellular processes and are essential modulators of responses to infection. Following our
identification of miR-19 as a positive regulator of NF-κB signalling, we investigated the role of this miRNA in the modulation of NF-κBdependent transcriptional responses. We identified a converging activity of miR-19 and NF-κB targeting, suggesting that their coordinate
activity regulates gene induction upon infection. Genome-wide microarray analyses following overexpression or repression of more than
60 miRNAs confirmed that in ~75% of cases, miRNA targets could be associated with the activity of a single over-represented
transcription factor. Collectively, our research demonstrates that miRNAs are master regulators of global transcriptional responses,
acting to dampen transcriptional variation or enforce transcriptional restriction. In addition to developing our latest findings on this topic, I
will discuss the far-reaching implications of these results pertaining to the field of interferon-driven transcriptional responses.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 86
254
Do G-CSF and Neutrophils Contribute to the Pathophysiology of Rheumatoid Arthritis?
Gabrielle Goldberg1, Simon Chatfield1, 2, Jane Murphy1, Ee Shan Pang1, Yunshun Chen1, Gordon Smyth1, Milica Ng3, Clare
O'Neill2, Samantha Busfield3, Arna Andrews3, Ian Wicks1, 2
1. Inflammation Division, Walter and Eliza Hall Institute, Parkville, Victoria, Australia
2. Rheumatology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia
3. CSL Limited, Bio21 Institute, Parkville, Victoria, Australia
Rheumatoid arthritis (RA) is characterised by a persistent, but poorly understood interplay between innate and adaptive immunity.
Neutrophils are the predominant cell type in inflamed RA synovial fluid (SF). Granulocyte colony-stimulating factor (G-CSF) is a key
regulator of neutrophil production, function and survival. In this study peripheral blood (PB) neutrophil phenotype and function were
analysed according to disease activity (DAS28 scores and other clinical parameters) in RA patients (n=50-60). We found significant
correlations between disease activity and PB neutrophil percentage (r=0.26, p≤0.05), as well as between neutrophil activation state
(CD62L, CD11b) and the expression of receptors for factors regulating neutrophil production and function. For example, CD62L vs GCSF-R (r=0.58, p≤0.0001); CD62L vs CD35 (r=-0.37, p≤0.01); CD62L vs CXCR2 (r=0.42, p≤0.01); CD62L vs CXCR1 (r=0.46, p≤0.001).
To further explore the role of neutrophils and G-CSF in RA, transcriptional profiling using RNASeq was performed comparing: PB
neutrophils isolated from healthy donors (HD) and RA patients (n=5); neutrophils isolated from paired PB and SF samples of RA
patients (n=3); neutrophils or white blood cells from HDs stimulated with G-CSF in vitro (n=4). 194 genes were differentially expressed
(DE) in RA neutrophils (logFC≥1; adjusted p≤0.05) when compared to HDs. Over 50 of those genes were also differentially expressed
when neutrophils were stimulated with G-CSF in vitro. There were 1724 DE genes (logFC≥1; adjusted p≤0.05) when comparing PB and
SF neutrophils from RA patients. Bioinformatic interrogation using Ingenuity Pathway Analysis software demonstrated that G-CSF was a
likely regulator (p=9.83x10-21) of these differences. These data provide evidence that neutrophils and G-CSF contribute to the
pathogenesis of RA. G-CSF may therefore represent a potential therapeutic target in the treatment of RA and other inflammatory
diseases where there is a pathogenic contribution from neutrophils.
255
G-CSF and Neutrophils are Non-Redundant Mediators of CNS Autoimmune Disease
Gabrielle L Goldberg1, Ann L Cornish1, Jane Murphy1, Ee Shan Pang1, Lyndell L Lim2, Ian Campbell3, Xiangting Chen4, Paul G
McMenamin4, Brent McKenzie3, Ian Wicks1, 5
1. Inflammation Division, Walter and Eliza Hall Institute, Parkville, Victoria, Australia
2. Centre for Eye Research Australia, University of Melbourne, Melbourne, Victoria, Australia
3. CSL Limited, Bio21 Institute, Parkville, Victoria, Australia
4. Department of Anatomy, Monash University, Clayton, Victoria, Australia
5. Rheumatology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia
Granulocyte colony stimulating factor (G-CSF) plays a key role in the regulation of neutrophil production, function and survival. The GCSF receptor (G-CSF-R) it most highly expressed by neutrophils and myeloid progenitors, and at lower levels by macrophages and
monocytes. Here, we investigated the role of G-CSF and neutrophils in two extensively used models of central nervous system (CNS)
autoimmunity both of which have long been described as T cell-mediated - experimental autoimmune encephalitis (EAE) and
experimental autoimmune uveoretinitis (EAU).In G-CSF-/- mice and in WT mice treated with anti-G-CSF monoclonal antibody (mAb),
disease severity in both models was dramatically reduced. A mixed population comprising macrophages ,T cells and neutrophils were
identified by flow cytometric analysis of the ocular infiltrate in WT mice with EAU. The eyes of G-CSF-deficient and anti-G-CSF mAbtreated WT mice had reduced disease severity, with markedly reduced neutrophil infiltrate, but little or no change in other myeloid
inflammatory cells. In the absence of G-CSF, antigen-specific T cell responses remained intact in both disease models and IL-17A
production was maintained. We demonstrate that G-CSF controls ocular neutrophil infiltrate by modulating the expression of chemokine
receptors - CXCR2 and CXCR4 - on peripheral blood neutrophils as well as CXCL2-mediated actin polymerization. These data establish
an integral role for G-CSF-driven neutrophil responses in CNS autoimmunity, operating both within and outside the bone marrow and
identify G-CSF as a potential therapeutic target in the treatment of human uveoretinitis and multiple sclerosis.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 87
256
The crystal structure of human interleukin-11: receptor binding site features and structural
differences from interleukin-6
Tracy Putoczki1, Renwick Dobson3, 2, Michael Griffin3
1. Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
2. Biomolecular Interactions Centre and School of Biological Sciences, University of Canterbury, Christchurch, New Zealand.
3. Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC,
Australia
Interleukin (IL)-11 is a member of the IL-6 family of pleiotropic cytokines that is characterised by shared use of the signal transducing
receptor, GP130. Similar to IL-6, signalling by IL-11 is initiated by binding of soluble IL-11 to its membrane bound, specific receptor, IL11Rα. This binary complex subsequently engages with GP130, inducing GP130 dimerisation and recruitment of JAK kinases, ultimately
resulting in phosphorylation and activation of the transcription factor, Signal Transducer and Activator of Transcription (STAT)-3. As a
result of the historical focus on the IL-6/STAT3 signalling axis, the IL-11 signalling machinery has remained relatively uncharacterised.
No high resolution structural data have previously been reported for IL-11 or IL-11Rα and our understanding of the structure of the
authentic signalling complex and the interactions between its components remains rudimentary. Here we report the crystal structure of
human IL-11 and provide structural resolution of residues previously identified as important for IL-11 activity. Biophysical analysis
indicates that IL-11 is a compact, stable, monomeric protein. While IL-11 is thought to signal via a complex analogous to that of IL-6, our
comparisons show important differences between the two cytokines. Specifically, IL-11 shows a more elongated structure than IL-6 and
lacks additional helical elements surrounding its central, 4-helix core. Detailed analysis of the GP130 binding regions suggest that IL-11
engages the first molecule of GP130 differently to IL-6. In addition to providing a structural platform for further study of IL-11, these data
offer insight into the binding interactions of IL-11 with each of its receptors and the structural mechanisms underlying agonist and
antagonist variants of the protein.
1.
Putoczki T.L., Dobson R.C.J., Griffin M.D.W. (2014) The crystal structure of human interleukin-11 reveals receptor binding site
features and structural differences from interleukin-6. Acta Crystallographica D. In Press doi:10.1107/S1399004714012267
257
Strawberry notch homolog 2 gene expression is increased in the brain during endotoxin-induced
systemic inflammation and is localized to numerous cells including astrocytes and microglia
Magdalena Grill1, 2, Taylor E. Syme1, Aline L. Noçon1, Iain L. Campbell1
1. School of Molecular Bioscience, University of Sydney, Sydney, NSW, Australia
2. Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
Aims: In an effort to better understand the function of the cytokine Interleukin (IL)-6 in the murine brain, our group identified the putative
DExD/H-box helicase strawberry notch homolog 2 (Sbno2) as a novel IL-6/glycoprotein 130 regulated target gene in murine astrocytes
and microglia in vitro. The aim of this study was to determine the regulation and cellular localization of Sbno2 gene expression during
neuroinflammatory conditions in vivo.
Methods: Upon endotoxin-induced systemic inflammation (2 mg/kg dual LPS injection, intra-peritoneal, 16 h plus 4 h or 24 h,
respectively) in mice, total RNA was isolated from the brain and Sbno2 mRNA levels were determined by RNase protection assay. From
a second set of animals, paraformaldehyde-fixed, paraffin-embedded mouse brains were sagitally cut and analysed by in situ
hybridization (ISH)/immuno-/histochemistry (IHC).
Results: Sbno2 mRNA levels were highly increased at 4 h and decreased thereafter, but still had elevated levels up to 24 h after the
second treatment. In ISH experiments, up-regulated Sbno2 mRNA was mainly co-localized with GFAP-positive astrocytes while lesser
numbers of selected tomato lectin-stained microglia and endothelial cells co-localized with Sbno2 mRNA as well.
Conclusions: The results of the current study complement our previous in vitro data and strengthen the notion of a role for Sbno2 in the
innate inflammatory response during host-defence.
Supported by NH&MRC grants 632754 and APP1047265 and Austrian Science Fund (FWF): J3081-B09.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 88
258
Interferon gamma deficiency and Epstein-Barr virus reactivation in the pathogenesis of chronic
lymphocytic leukemia
Ewelina Grywalska1, Marcin Pasiarski, Agata Surdacka, Jacek Rolinski
1. Medical University of Lublin, Lublin, Poland
Aims: Interferon (IFN)-gamma is a crucial regulatory cytokine in cellular immunity, and is important in immune surveillance of EpsteinBarr virus. Pathogenesis of chronic lymphocytic leukemia (CLL) is associated with an array of not completely understood disorders of
cell- and humoral-mediated immunity. The aim of the study was an assessment of plasma concentration and intracellular expression of
IFN-gamma, and its relationship with Epstein-Barr virus (EBV) DNA copy number, and selected clinical parameters in patients with CLL
and healthy controls.
Methods: The study included samples of peripheral blood from 110 untreated patients with CLL. The control group comprised of 40
age- and sex-matched healthy individuals. The EBV-DNA copy number in peripheral blood mononuclear cells (PBMCs) was analyzed
with EBV PCR kit (a single gene encoding EBV nuclear antigen 1 was amplified). The immunophenotype of PBMCs, including
intracellular expression of IFN-gamma, was determined with flow cytometer. Concentration of INF-gamma in plasma was determined by
ELISA.
Results: Presence of EBV DNA in PBMCs was found in 59 patients with CLL. There was a lack of detectable amounts of viral genetic
material in healthy individuals. Decreased number of lymphocytes showing intracellular expression of IFN-gamma (CD3+/CD4+/IFNgamma+ T cells, p=0.0027; CD3+/CD8+/IFN-gamma+ T cells, p=0.0044; CD19+/IFN-gamma+ B cells, p=0.0089), and low plasma
concentration (p<0.0001) of this cytokine in CLL EBV(+) patients, as compared to EBV(-) subjects and healthy controls, reflect the
suppression of cell-mediated immune response and non-reactivity of lymphocytes to the studied virus. The most important factors
shortening the survival of CLL patients and time to progression were: EBV-DNA copy number (>17 copies/ug DNA) and low plasma
concentration of IFN-gamma (<5.25 pg/ml).
Conclusion: These data show IFN-gamma deficiency in some patients with CLL and suggest that normal T- and B-cell response is
crucial for favorable prognosis in this disease.
Acknowledgments: This work was supported by research grants: NN402682440, 2011/01/N/NZ6/01762 and 2012/05/B/NZ6/00792 from
State Funds for Scientific Research National Science Centre (NCN, Poland).
259
Using Type III Interferon to Enhance Oncolytic Virus Function
Ryann C. Guayasamin1, Michael D. Robek1
1. Yale School of Medicine, New Haven, CT, United States
Introduction
Hepatocellular carcinoma (HCC) is responsible for more than 500,000 yearly deaths worldwide. The lack of effective therapies for this
disease necessitates novel therapeutic approaches. Oncolytic viral vectors, such as vesicular stomatitis virus (VSV), are a promising
new anticancer platform for HCC. In addition to direct cytopathology, VSV infection of cancer cells elicits specific and nonspecific
immune responses, thus overcoming immunological tolerance to the tumor. The type III IFN family of cytokines (IFN-λ1, 2, and 3) have
antiviral and immunomodulatory properties, and inhibit tumor progression in several different cancer models. We hypothesize that IFN-λ
expression from VSV will improve oncolytic activity by both immunostimulatory effects and through increased selectivity of the virus for
tumor cells.
Methods
We generated recombinant VSV vectors expressing mouse IFN-l2 either from the fifth position or the more highly expressing first
position of the viral genome, and measured virus replication in cell culture and mice. Using a mouse model of HCC, we are examining
the hypothesis that IFN-l expression from VSV will augment its antitumor activity.
Results
We found that IFN-l is expressed, secreted, and active when produced by VSV-infected cells. In cell culture, IFN-l expression from VSV
attenuated virus replication in type III IFN-responsive cells, but not in type III IFN-nonresponsive cells, and after intranasal delivery to
mice in vivo (Guayasamin et al., J. Virol., 2014). In vitro virus infection and spread assays demonstrate that VSV vectors expressing
IFN-l are selectively cytopathic for type III IFN-nonresponsive liver cancer cells but not normal hepatocytes. Mouse models of HCC are
being used to determine the antitumor potency of the VSV vectors and are ongoing.
Conclusion
These studies demonstrate the ability of the type III interferon cytokine family to enhance VSV oncolytic activity by increasing selectivity
of the virus for type III IFN-resistant tumor cells.
Conclusions/Significance: We suggest that B. pseudomallei-infected CD11b+CD62L+ cells play an important role in inducing melioidosis
with meningitis that could be due to the migration of infected leukocyte involving in a selectin-dependent manner.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 89
260
Functional Promiscuity in C-terminal Peptidic C3aR/C5a1 Ligands
Reena Halai1, Meghan L Bellows-Peterson2, Will Branchett3, James Smadbeck2, Chris A Kieslich2, Daniel E Croker1, Matt A
Cooper1, Dimitrios Morikis4, Trent M Woodruff5, Christodoulos A Floudas2, Peter N Monk3
1. Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
2. Department of Chemical and Biological Engineering, Princeton University, NJ, USA
3. Department of Infection and Immunity, Sheffield University Medical School, Sheffield, UK
4. Department of Bioengineering, University of California, Riverside, CA, USA
5. School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
Background: The complement cascade is a highly sophisticated network of proteins that are well regulated and directed in response to
invading pathogens or tissue injury. Complement C3a and C5a are key mediators produced by this cascade, and their dysregulation has
been linked to a plethora of inflammatory and autoimmune diseases. Consequently, this has stimulated interest in the development of
ligands for the receptors for these complement peptides, C3aR, and C5a1 (C5aR/CD88).
Aims: In this study we used computational methods to design novel C5a1 receptor ligands.
Results: Functional screening in HMDM using the xCELLigence label-free platform demonstrated promiscuity in the specificity of our
ligands. No agonist/antagonist activity was observed at C5a1, but we instead saw that some ligands were able to agonize the closely
related complement receptor C3aR. This was verified in the presence of C3aR antagonist SB 290157 and in a stable cell line
expressing either C5a1 or C3aR alone.
Conclusions: C3a agonism has been suggested to be a potential treatment of acute neutrophil-driven traumatic pathologies, and may
have great potential as a therapeutic avenue in this arena.
261
Shifts in the microbiome during experimental chronic obstructive pulmonary disease (COPD)
Phil Hansbro1
1. University of Newcastle, Callaghan, NSW, Australia
Chronic obstructive pulmonary disease is the 3rd commonest in the world. It is a heterogeneous disease characterised clinically by
difficulties in breathing. It is induced by cigarette smoking that causes airway inflammation. Chronic inflammation results in tissue
damage leading to airway remodelling and fibrosis, alveolar destruction and emphysema that combine to impair lung function. Current
therapies alleviate some symptoms but do not reverse nor cure the disease. The microbiome encompasses all the microorganisms that
inhabit the body. Recent studies show that a healthy microbiome is essential for maintaining homeostasis and health. Alterations in
microbiomes in various tissues (eg gut) is associated with disease (eg colitis), potentially by inducing local and systemic inflammation.
These changes may be reversed by microbiome transfer (eg colitis). Other recent studies have demonstrated that substantial immune
and inflammatory cross-talk occurs between the lung and gut. We investigated the changes that occur in the gut microbiome in a mouse
model of COPD that we developed. We used microbiome profiling to identify substantial shifts in microbial populations during the
induction and progression of COPD. Transfer of fecal microbiomes from smoking groups to air groups and vice versa was performed,
with transfer resulting from coprophagy. This suppressed the development of COPD in smoke-exposed mice exposed to the faeces of
non-smoking mice. Profiling identified 2 species associated only with COPD and 2 species associated with protection. Analysis to
identify genus-level shifts or key microorganisms that cluster with diseased or healthy states is ongoing.
262
Autophagic Regulation of Pro-inflammatory Cytokines
Nichita Gavrilescu1, Tali Lang1, Jacinta Lee1, Andrew Foote1, Sarah Jones1, Eric Morand1, James Harris1
1. Centre for Inflammatory Diseases, Monash University, Clayton, Victoria, Australia
Autophagy is a conserved homeostatic mechanism for the lysosomal degradation of cytosolic constituents, including long-lived
macromolecules, organelles and intracellular pathogens. Autophagosomes are formed in response to a number of environmental
stimuli, including amino acid deprivation, but also by both host- and pathogen-derived molecules, including Toll-like receptor ligands and
cytokines. In turn, autophagy regulates the production and secretion of a number of cytokines. In particular, inhibition of autophagy in
LPS-treated macrophages and dendritic cells leads to increased secretion of IL-1α, IL-1β, IL-18 and IL-23. The mechanisms underlying
these effects differ between cytokines; secretion of IL-1β is dependent on activation of inflammasomes and the TLR2/TLR4 adaptor
molecule TRIF, while IL-1α secretion in response to autophagy inhibition is dependent on neither. Moreover, stimulation of autophagy
leads to a decrease in intracellular levels of pro-IL-1β, while the secretion of IL-23 in response to inhibition of autophagy is dependent on
IL-1. Moreover, supernatants from LPS-stimulated, autophagy-deficient, antigen presenting cells induce the secretion of IL-17, IL-22 and
IFN-γ by γδ+ T cells, suggesting that autophagy indirectly regulates Th17 responses through the modulation of IL-1 and IL-23. Thus,
autophagy represents a potential target for the development of novel therapeutics for the treatment of inflammatory and autoimmune
disorders, as well as in the development of treatment strategies for certain infectious diseases and vaccines.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 90
263
Antagonism of STAT3 signaling by lyssaviruses: a potent mechanism to inhibit cytokine responses,
and potential target for antivirals
Angela Harrison, Kim Lieu, Aaron Brice, Gregory Moseley
Lyssaviruses, including rabies virus (RABV), are the etiological agents of rabies, an incurable disease with a case fatality rate of 100%
that causes >60,000 human deaths/year. In common with other human-pathogenic viruses, RABV infection depends on evasion of host
interferon (IFN)-mediated immunity, which is mediated through the interaction of the RABV IFN-antagonist P-protein with IFNactivated signal transducers and activators of transcription (STATs) 1 and 2, which has been shown to be critical to disease
progression in vivo. Intriguingly, recent data indicate that RABV P-protein also targets STAT3, a major mediator of signaling by IFNs and
by members of the IL-6 cytokine family. Similar reports for IFN-antagonists of several paramyxoviruses indicate that STAT3 targeting
might play critical roles in infection by many viruses. However the molecular details underlying STAT3 targeting, and outcomes in terms
of cytokine signaling and disease, remain largely unresolved.
Using quantitative cell imaging, protein interaction analysis and immune signaling assays, we have analysed STAT3 targeting by Pproteins from a panel of diverse lyssaviruses, finding that P-protein inhibits STAT3 responses to cytokines of BOTH the IFN and IL-6
families, and that STAT3-targeting is highly conserved across the genus, indicative of important roles. Nevertheless, we also identified
subtle but significant differences in STAT3 targeting between different lyssaviruses, and identified specific mutations of P-protein able to
prevent STAT3 interaction. Importantly, these mutations also reduce pathogenicity in mice. Using these tools, we are currently
determining the precise molecular basis of P-protein’s interaction with STAT3, and the roles of P-protein-STAT3 complexes in
modulating antiviral immune signaling. This work should significantly enhance our understanding of the interaction of RABV with multiple
cytokine signaling pathways, to reveal the extent of viral interference in immune signaling and its importance to disease, identifying new
strategies to combat currently incurable rabies disease.
264
G-CSF is a critical mediator of chronic inflammatory lung disease
Evelyn Tsantikos1, Mhairi J Maxwell1, Maverick Lau1, 2, Erika Duan1, Gary P Anderson2, Margaret L Hibbs1
1. Department of Immunology , Monash University, Melbourne, VIC, Australia
2. Lung Health Centre, University of Melbourne, Melbourne, VIC, Australia
Chronic obstructive pulmonary disease (COPD) is a health problem of utmost global significance affecting tens of millions of people and
it is resistant to current therapies. COPD is hallmarked by an irreversible and progressive deterioration in lung function, which is caused
by destruction of lung parenchyma due to inflammation, and fibrosis in small airways. Most COPD is caused by irritants (smoke) and
one near universal feature of COPD is a heightened chronic inflammatory response in lung involving macrophages, epithelial cells,
granulocytes, dendritic cells and lymphocytes. Lung injury caused by cigarette smoke and irritants strongly induces the release of many
inflammatory mediators including myeloid colony-stimulating factors (CSFs). CSFs link development of granulocytes and macrophages
from the bone marrow with tissue inflammation, autoimmunity and host defence, and while GM-CSF is highly implicated in COPD
pathogenesis, the role of G-CSF is much less well understood. The SH2 domain-containing inositol 5’ phosphatase SHIP-1 is a negative
regulator of myeloid cell proliferation and growth-factor mediated survival, exemplified in mice where deletion of SHIP-1 causes
neutrophil and myeloid cell hyperplasia, myeloid cell accumulation in the spleen, lymph nodes and lungs, and the development of
emphysema and spontaneous lung fibrosis that resembles human COPD. G-CSF levels are significantly elevated in the serum and
bronchoalveolar lavage fluid of SHIP-1-deficient mice and the mice exhibit dramatically increased numbers of myeloid progenitors in
spleen. We now show that deletion of G-CSF from SHIP-1-deficient mice leads to their increased survival, and the mice exhibit
markedly reduced lung disease, loss of splenomegaly and lymphadenopathy phenotypes, and dramatic diminution of extramedullary
hematopoiesis. Collectively, our findings show that G-CSF plays a crucial role in mobilizing hematopoietic stem cells from the bone
marrow and inciting tissue inflammation in SHIP-deficient mice, which plays an important role in the development of chronic
inflammatory lung disease.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 91
265
Human airway epithelial cells express ABCC4/MRP4, a transporter for uric acid and cAMP secretion,
that contributes to innate and adaptive immune responses.
Jeremy A Hirota1, Matt Gold, Paul Hiebert, HyeYun Park, Dorota Stefanowicz, Kelly McNagny, Darryl Knight, Don Sin, Chris
Carlsten
1. University of British Columbia, Vancouver, BC, Canada
HYPOTHESIS: Uric acid can be detected in airway lining fluids, although the mechanism regulating this process remains unclear. Uric
acid is a potent anti-oxidant at physiological concentrations while becoming a danger associated molecular pattern triggering innate
immune responses at pathological concentrations. Extracellular cAMP has also been shown to modulate immune responses. We
tested the hypothesis that human airway epithelium expresses ABCC4/MRP4 which can regulate uric acid and cAMP secretion
contributing to context dependent airway health and pathology.
METHODS: We used in vitro experiments with human airway epithelial cells and in vivo mouse models of airway disease. We
performed candidate uric acid transporter gene expression, in situ immunohistochemistry, and in vitro pharmacological intervention
experiments with human airway epithelial cells. We assessed the functional consequences of this system in a mouse model of allergic
airways disease.
RESULTS: We demonstrate gene and protein expression of ABCC4/MRP4 in human airway epithelial cells in vitro and in situ. In
vitro airway epithelial cells secrete basal levels of uric acid further induced with allergen treatment. ABCC4/MRP4 inhibition significantly
blocked basal and induced uric acid secretion, without changes in cell death. This data suggests a uric acid transport system is active
and does not require cell death. Inhibition of ABCC4/MRP4 also attenuated forskolin induced cAMP secretion. In vivo intervention
studies targeting uric acid at the mucosal surface prevented allergic sensitization in a model of air pollution and allergen co-exposure.
CONCLUSIONS: Our results are the first to demonstrate the presence and function of a uric acid and cAMP transport system in human
airway epithelial cells. Our results open the door to begin exploring how this transport system is regulated in health and disease, the
downstream consequences of activation of the system, and the potential for the system to be active at other mucosal surfaces
throughout the body.
Research Funding Source: Canadian Institutes of Health Research. BC Lung Association. Canadian Banting Fellowship Program
266
IFN-lambda production of CD8 cDCs in response to DNA-viruses, transfected dsDNA or cyclic-dinucleotides depends on STING
Henning Lauterbach1, Barbara Bathke1, Paul Chaplin1, Mark Suter2, Meredith O'Keeffe3, Hubertus Hochrein1
1. Bavarian Nordic, Martinsried, Germany
2. University of Zurich, 8006 Zurich, Switzerland
3. Centre for Biomedical Research, Burnet Institute, Melbourne, Vic, Australia
The CD8+ conventional dendritic cells (CD8 cDCs) and the plasmacytoid dendritic cells (pDC) are major producers of IFN-lambda (IL28/29). Previously we have shown that upon stimulation with dsRNA the murine CD8 cDCs were the exclusive source of IFN-lambda in
vivo and in vitro via a TLR3/TRIF dependent recognition pathway. A finding confirmed for the human equivalent cDCs expressing
CD141 (BDCA3). In contrast, pDCs ignored dsRNA but produced large amounts of IFN-lambda in response to TLR7 or TLR9 stimuli in
a MyD88 dependent way. Here we demonstrate that both, the CD8 cDCs and pDCs, are able to produce IFN-lambda in response to
DNA viruses such as HSV-1 or poxviruses. Our analysis of the pattern recognition receptors or adaptor molecules involved revealed
that pDCs mainly use TLR9 and MyD88 for the detection of the DNA viruses, whereas CD8 cDCs depend on the adaptor molecule
STING but not on MyD88. The transfection of CD8 cDCs with different forms of dsDNA or cyclic-di-nucleotides such as c-di-GMP or cGAMP induced, similar to the infection with DNA viruses, large amounts of IFN-lambda in a STING dependent way. Thus, the immune
system produces IFN-lambda in response to DNA via two different DC subsets (CD8 cDCs and pDCs), different pattern recognition
receptors (cytoplasmic DNA receptors and TLR9) and adaptor molecules (STING and MyD88) as well as downstream signalling
components (IRF3 and IRF7). We hypothesize; that this widespread redundancy counteracts viral or other pathogen encoded inhibitory
mechanisms, possibly acting on the specific DC subsets, the pattern recognition receptors and adaptor molecules or other signalling
components.
267
Proteomic analysis of mitochondrial-associated ER membranes during RNA virus infection reveals
dynamic changes in protein and organelle trafficking
Stacy M Horner1
1. Duke University Medical Center, Durham, NC, United States
RIG-I pathway signaling of innate immunity to RNA virus infection is organized between the ER and mitochondria on a subdomain of the
ER called the mitochondrial-associated ER membrane (MAM). The RIG-I adaptor protein MAVS transmits downstream signaling of
antiviral immunity, with signaling complexes assembling on the MAM in association with mitochondria and peroxisomes. To identify
components that regulate MAVS signalosome assembly on the MAM, we characterized the proteome of MAM, ER, and cytosol from
cells infected with either chronic (hepatitis C) or acute (Sendai) RNA virus infections, as well as mock-infected cells. Comparative
analysis of protein trafficking dynamics during both chronic and acute viral infection reveals differential protein profiles in the MAM
during RIG-I pathway activation. We identified proteins and biochemical pathways recruited into and out of the MAM in both chronic and
acute RNA viral infections, representing proteins that drive immunity and/or regulate viral replication. In addition, by using this
proteomics approach, we identified 3 new MAVS-interacting proteins, RAB1B, VTN, and LONP1. Our proteomic analysis also reveals a
dynamic cross-talk between subcellular compartments during both acute and chronic RNA virus infection, and demonstrates the
importance of the MAM as a central platform that coordinates innate immune signaling to initiate immunity against RNA virus infection.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 92
268
Identification and functional characterization of K+ transporters encoded by Legionella pneumophila
kup genes.
Juliana I. Hori1, Marcelo M.S. Pereira1, Craig R. Roy2, Hiroki Nagai3, Dario S. Zamboni1
1. FMRP/USP, Ribeirão Preto, SP, Brazil
2. Section of Microbial Pathogenesis, Yale University, New Haven, CT, USA
3. Osaka University, Osaka, Japan
Legionnaires' disease is an emerging, severe, pneumonia-like illness caused by the Gram-negative intracellular bacteria Legionella
pneumophila, which are able to infect and replicate intracellularly in macrophages. Little is known regarding the mechanisms used by
intracellular L. pneumophila for the acquisition of specific nutrients that are essential for bacterial replication. Here, we investigate
three L. pneumophila genes with high similarity to the Escherichia coli K+ transporters. These three genes were expressed by L.
pneumophila and have been designated kupA, kupB and kupC. Investigation using the L. pneumophila kup mutants revealed
that kupA is involved in K+ acquisition during axenic growth. The kupA mutants replicated efficiently in rich axenic media, but poorly in a
chemically defined medium. The kupA mutants were defective in the recruitment of polyubiquitinated proteins to the Legionellacontaining vacuole that is formed in macrophages and displayed an intracellular multiplication defect during the replication
in Acanthamoeba castellanii and in mouse macrophages. We found that bafilomycin treatment of macrophages was able to rescue the
growth defects of kupA mutants, but it did not influence the replication of wild-type bacteria. These defects identified in kupA mutants
of L. pneumophila were complemented by the expression E. coli trkD/kup gene in trans, a bona fide K+ transporter encoded by E.
coli. Finally, we observed that the kupA mutant failed to activate the caspase-1 and also showed a reduction in the secretion of the IL-1β
cytokine. Collectively, our data indicate that KupA is a functional K+ transporter expressed by L. pneumophila that facilitates the
bacterial replication intracellularly and in nutrient-limited conditions. However, KupA can also be involved with the activation of the
inflammasomes.
269
Examination of inflammatory pathway induction in the fruit bat.
aaron T irving1, Matae Ahn1, Linfa F Wang1, 2
1. Duke-NUS Graduate Medical School, Singapore, Singapore
2. Australian Animal Health Laboratory, CSIRO Livestock Industries, East Geelong, Victoria, Australia
Bats are known to be the natural reservoir hosts of multiple pathogenic viruses with frequent zoonotic spillover events into human
populations. However, most virus infections are completely asymptomatic in bats. These same virus infections cause massinflammation in animals and humans. We are investigating how pathogen detection and activation of the Interferon signaling pathways,
and cross talk to the inflammasome pathway may be altered in the Australian black flying fox. Our data so far suggests that while bats
can mount a sufficient, yet altered, induction of the IFN pathway with production of IFN α, β and λ and various ISGs, and induction of
NFκB signaling, most pathogen detection does not result in activation of the inflammasome signaling complexes. Interrogation of the
inflammasome pathways reveals poor Asc-recruitment and low caspase activation in response to natural and synthetic ligands. Whether
this is due to altered kinetics, a decrease in sensitivity or redundancy of signaling pathways is currently under investigation.
270
Regulation of cell survival signals by Interleukin-3 in myeloid cells
Dimitra Masouras1, Carmel Daunt1, Karla Fischer1, Jarrod Sandow1, Paul Ekert2, Anissa Jabbour1
1. Walter + Eliza Hall Institute, Parkville, VIC, Australia
2. Murdoch Children's Research Institute, Melbourne
Survival and proliferation of haematopoietic cells is tightly regulated by cytokines, such as Interleukin (IL)-3. When IL-3 is not available,
dependent cells arrest in G1 and activate intrinsic apoptosis pathways. The Bcl-2 family of proteins regulates intrinsic cell death in
response to IL-3 loss. We have previously shown that deletion of the Bcl-2 family member, a BH3-only protein Puma, in myeloid cells
affords protection from Interleukin-3 (IL-3) withdrawal mediated apoptosis. In the absence of IL-3, Puma protein levels increase in a p53dependent manner. If IL-3 is restored before cells commit to apoptosis, Puma levels decrease in a manner dependent on posttranslational phosphorylation of Puma at serine 10 by IKK1. These results indicate a role for the IKK complex downstream of IL-3
signalling. Using myeloid cells lacking IKK1 or IKK2 we show the relevance of IKK dependent survival signals downstream of the IL-3
receptor.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 93
271
An increased level of IL-6 suppresses NK cell activity in peritoneal fluid of patients with
endometriosis via regulation of SHP-2 expression
Haiyoung Jung1, 2, Young-Ju Kang1, Arum Park1, 2, Young-Jun Park1, 2, Tae-Don Kim1, 2, Inpyo Choi1, 2, Suk Ran Yoon1, 2
1. KRIBB, 125 Gwahak-ro, Yuseong-gu, South Korea
2. Functional Genomics, UST, Deajeon, South Korea
Endometriosis is known to be related to a defect in NK cell cytolytic activity. Additionally, the levels of inflammatory cytokines are
elevated in the peritoneal fluid (PF) of women with endometriosis. Here, we show the decrease of natural killer (NK) cell cytolytic activity
in the PF of endometriosis patients by primary cytokine activity. An increased level of interleukin-6 (IL-6) in the PF of patients with
endometriosis suppresses NK cell cytolytic activity by down-regulating cytolytic granule components, such as granzyme B and perforin,
through the modulation of Src homology region 2-containing protein tyrosine phosphatase-2 (SHP-2) expression, suggesting that IL-6
plays a crucial role in the reduction of NK cell activity in the PF of patients with endometriosis.
272
Functional expression of umami taste receptor T1R1/T1R3 in mouse neutrophils
Young Su Jung1, NaHye Lee2, Ha Young Lee3, 1, Yoo jung Park1, JaeHyung Koo2, Yoe-Sik Bae3, 4, 1
1. Department of Biological Sciences, Sungkyunkwan University, Suwon
2. Daegu Gyeonbuk Institute of Science and Technology (DGIST), Daegu
3. Mitochondria Hub Regulation Center, Dong-A University, Busan
4. Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul
Although many different types of G-protein coupled receptors (GPCRs) are functionally expressed in neutrophils, no reports have
demonstrated functional expression of umami taste receptor in these cells. In this study, we observed that mouse neutrophils express
the umami taste receptor heterodimeric T1R1/T1R3 through quantitative RT-PCR analysis. We also found that stimulation of mouse
neutrophils with L-alanine or L-serine, which is a ligand for the umami taste receptor, elicited not only ERK or p38 MAPK
phosphorylation but also neutrophil chemotactic migration. Moreover, addition of L-alanine or L-serine markedly blocked the production
of several cytokines including TNF-α, CCL2, and IL-10 induced by lipopolysaccharide through inhibition of NF-κB activity or STAT3
phosphorylation in neutrophils. Our findings demonstrate that neutrophils express the umami taste receptor, and tastants stimulate
neutrophils, resulting in chemotactic migration, and attenuation of LPS-induced inflammatory response.
273
Interferon-induced autophagy and antibacterial defenses occur through the Apoptosis stimulating
kinase-1
Dhan V Kalvakolanu1, Padmaja Gade1, Srikantha B Manjegowda1, Shreeram C Nallar1
1. University of Maryland Baltimore, Baltimore, MARYL, United States
IFN-g induces multiple gene regulatory pathways to combat pathogens and tumors. The death-associated protein kinase 1 (DAPK1) is
an important IFN-induced regulator of cell death and autophagy. Recently we have identified that ATF6, an ER resident transcription
factor, in association with the transcription factor CEBP-b regulates the IFN-g-induced expression of Dapk1 though a novel enhancer
element. ATF6 migrates to Golgi in response to IFN-g, where it undergoes proteolysis to yield the transcriptionally active form that
enters nucleus and induces gene expression. IFN-g-induced proteolytic processing of ATF6 and phosphorylation of C/EBP-b were
essential for the formation of a novel transcriptional complex that regulates Dapk1. ASK1 (MAP3K5) is activated by various stress
stimuli. Although originally identified as a kinase that stimulates apoptosis, ASK1 also contributes to cytokine responses, cell
differentiation and immunity. However, the exact targets of this kinase are unclear. Here, we report that IFN-γ activates ASK1MKK3/MKK6-p38MAP kinase pathway for controlling the activity of ATF6. The terminal enzyme in this pathway, p38MAP kinase,
phosphorylates a critical threonine residue in ATF6, upstream of its DNA binding domain, which is required for its proteolytic processing.
ATF6 mutants lacking the p38 MAPK phosphorylation site fail to undergo proteolytic processing in the Golgi, drive IFN-g induced gene
expression and autophagy. We also show that mice lacking Ask1 are highly susceptible to lethal bacterial infection owing to defective
autophagy. Together, these results identify a novel host-defense pathway controlled by IFN-g.
274
Cytokine-induced tumor suppression: a GRIM lesson
Dhan Kalvakolanu1, Sudhakar Kalakonda1, Shreeram C Nallar
1. University of Maryland Baltimore, Baltimore, MARYL, United States
GRIM-19, a STAT3 inhibitory protein, was isolated as a growth suppressive gene product using a genome-wide expression knockdown
screen as an Interferon/retinoid induced growth suppressors. Loss of expression and occurrence of mutations in the GRIM-19 gene in a
variety of primary human cancers, suggesting its importance as a novel tumor suppressor. A number of DNA viral and cellular
oncogenes are blocked by GRIM-19. We generated a Grim-19 conditional knockout mouse for understanding its tumor suppressor
function in vivo. Deletion of Grim-19 significantly increased susceptibility of mice to chemical carcinogenesis resulting in development of
squamous cell carcinomas. These tumors had high STAT3 activity and an increased expression of STAT3 responsive genes.
Surprisingly, mono-allelic loss of Grim-19 gene was sufficient to promote carcinogen-induced formation of invasive squamous cell
carcinomas. Loss of Grim-19 also caused mitochondrial electron transport dysfunction and assembly of the ETC complexes, and altered
the expression of several cellular genes involved in glycolysis and promoted Warburg effect. These observations highlight the critical
role of GRIM-19 as a cytokine-induced tumor suppressor.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 94
275
Dectin-1 modifies colonic microflora by inducing cytokine-dependent antimicrobial peptide secretion
from intestinal epithelial cells.
Tomonori Kamiya1, Tange Ce1, Yoichiro Iwakura1
1. Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, CHIB, Japan
The mucosal immune system regulates the balance of intestinal microflora by innate immunological molecules such as antimicrobial
peptides (AMPs). Dectin-1 is the receptor for β-glucans which are a component of fungal cell walls and are contained in various foods,
suggesting that Dectin-1 signaling may regulate intestinal mucosal immunity. Here, we found that calprotectin, one of AMPs, was
exclusively produced by mouse colonic epithelial cell (cEC), and Dectin-1 deficient mice showed significantly impaired calprotectin
expression in cEC. Calprotectin was induced by a special IL-17 family cytokine that was mainly produced by macrophages in colonic
lamina propria. Calprotectin specifically suppressed the growth of a species of intestinal commensal bacteria belonging to Firmicutes,
and the population of this species was increased in both Dectin-1 and this IL-17 family cytokine deficient mice. Furthermore, the ligand
of Dectin-1 in mouse intestine was identified as food-derived β-glucans. These findings suggest that oral administration of β-glucans
regulates the balance of commensal bacteria population via the Dectin-1-induced cytokine-dependent AMP secretion to control the
homeostasis of intestinal immunity.
276
The IL-3 receptor α subunit N-terminal domain: role in receptor assembly and signalling
Winnie L Kan1, Timothy R Hercus1, Sophie E Broughton2, Tracy L Nero2, Mara Dottore1, Frank C Stomski1, Emma F Barry1,
Barbara J McClure1, Urmi Dhagat2, Matthew P Hardy3, Nicholas J Wilson3, Michael W Parker2, Angel F Lopez1
1. Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
2. ACRF Rational Drug Discovery Centre, St Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
3. CSL Limited, Parkville, VIC, Australia
Interleukin (IL)-3, granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-5 are responsible for the survival, proliferation,
differentiation and activation of hematopoietic cells. IL-3 signals through a receptor consisting of a ligand-specific α subunit (IL3Rα) and
a β subunit (βc), which is shared with the GM-CSF and IL-5 receptors. IL-3Rα is overexpressed on the surface of leukaemic stem cells
in patients with acute myelogenous leukaemia, allowing the targeting of leukaemic stem cells with specific antibodies 1. Our previous
studies demonstrated that the GM-CSF receptor assembles into a hexamer consisting of 2 GM-CSF molecules, 2 αsubunits and a βc
homodimer, which subsequently forms a higher-order dodecamer complex upon cytokine binding that is required for Jak2
transphosphorylation and full activation2. We have recently solved the structure of IL3Rα, which contains the N-terminal domain (NTD)
and domains 2 and 3 and demonstrated that IL-3 can bind IL-3Rα through “open” and “closed” conformations of the receptor3. What
remains unknown is how the NTD contributes to receptor signalling and its functional significance.
The aim of the present study is to determine the role of the NTD of IL-3Rα in IL-3 binding, higher-order receptor assembly and cell
signalling. We generated several mutants that disrupt the conformation of the NTD, which were tested in signalling and functional
studies. The results showed that the binding affinity of IL-3 to the mutant receptors and proliferative activity in immortalized mouse fetal
liver cells expressing the mutants were reduced and similar to that observed for IL-3Rα that lacks the NTD. We will present further
biochemical data that suggests that the mutants follow different pathways to the formation of the receptor dodecameric complex.
Although the IL-3Rα NTD is not essential for ligand binding, its conformation once ligand-bound plays a crucial role in initiating and
directing the assembly of a functional higher order complex.
1.
2.
3.
Jin, L et al. (2009). Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid
leukemic stem cells. Cell Stem Cell 5, 31-42.
Hansen G et al. (2008). The structure of the GM-CSF receptor complex reveals a distinct mode of receptor activation. Cell 134,
496-507.
Broughton, SE et al. (2014). Dual Mechanism of interleukin-3 receptor blockade by an anti-cancer antibody. Cell Reports 8, 410419.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 95
277
Re-Zincing the macrophage antimicrobial response to Salmonella typhimurium.
Ronan Kapetanovic1, Nilesh J Bokil1, Maud ES Achard2, Kate Peters2, Minh Duy Phan2, Han Rong Foo1, Nick A Hamilton1, Kate
Schroder1, Katharine M Irvine3, Bernadette M Saunders4, Kate J Stacey2, Alastair G McEwan2, Mark A Schembri2, Matthew J
Sweet1
1. Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, The University of Queensland , St Lucia,
QLD, Australia
2. School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of
Queensland, St Lucia, QLD, Australia
3. School of Medicine, The University of Queensland, Wooloongabba, QLD, Australia
4. Mycobacterial Research Program, Centenary Institute, Locked Bag No. 6, Sydney, NSW, Australia
Zinc deficiency predisposes to infectious diseases and zinc supplementation is associated with improved outcomes in severe diarrheal
disease. Cytokines, including TNF and IL-6, released during inflammatory events induce an acute redistribution of the zinc, leading to
plasma zinc depletion. Here we investigated potential antimicrobial effects of zinc in human macrophages.
We show Toll-like Receptor (TLR) signalling promotes the delayed accumulation of vesicular zinc within primary human macrophages.
Treatment of macrophages with exogenous zinc does not alter LPS-dependent TNF and IL-6 production but increased the intracellular
zinc pool. After infection, the vesicular zinc is delivered to non-pathogenic Escherichia coli (E. coli) strain MG1655 for microbial
clearance. In contrast, Salmonella enterica serovar Typhimurium (S. Typhimurium) evades this response via a mechanism dependent
upon Salmonella pathogenicity island-1 (SPI-1); a SPI-1 mutant, but not wild type S. Typhimurium, co-localized with zinc-containing
vesicles. The antimicrobial effect of exogenous zinc was more pronounced with E. coli than with S. Typhimurium, suggesting that S.
Typhimurium may employ additional mechanisms to evade zinc-mediated antimicrobial responses. Indeed, intramacrophage S.
Typhimurium upregulated expression of the zinc exporter zntA, which effluxes zinc when concentrations reach cytotoxic levels. The
SPI1 mutant displayed prolonged up-regulation of zntA within macrophages, consistent with its sustained exposure to zinc in this
environment. Interestingly, Salmonella zntA mRNA is upregulated by both zinc and copper, and both metals ions colocalise in vesicles
in TLR-activated macrophages. Furthermore, copper and zinc synergized to reduce bacterial growth of the Salmonella zntA mutant.
Our data thus suggest that S. Typhimurium employs multiple mechanisms of zinc evasion including SPI-1-dependent subversion of zinc
delivery, upregulation of zntA and an additional yet to be characterized mechanism. In total, our findings identify roles for zinc and zinc
trafficking in human macrophage antimicrobial pathways against Gram-negative bacteria, and provide insights into host subversion.
278
Suppressor of cytokine signalling (SOCS) 5 regulates the innate anti-viral responses to influenza A
infection.
Lukasz Kedzierski1, 2, Michelle Tate3, Edmond Linossi1, 2, Sarah Freeman1, 2, Nicola Bird4, Bridie Day4, Tatiana Kolesnik1, 2,
Gabrielle Belz5, 2, Benjamin Kile6, 2, Nicos Nicola7, 2, Katherine Kedzierska4, Sandra Nicholson1, 2
1. Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
2. Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
3. Centre for Innate Immunity and Infectious Disease , MIMR-PHI Institute of Medical Research, Clayton, VIC, Australia
4. Department of Microbiology and Immunology, University of Melbourne At Peter Doherty Institute for Infection and Immunity,
Parkville, VIC, Australia
5. Molecular Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
6. Chemical Biology Division , Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
7. Cancer and Haematology Division , Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
The suppressors of cytokine signalling (SOCS) proteins are negative regulators of cytokine signalling, immune cell development and
function, and the inflammatory response. SOCS1-3 and CIS have defined roles in controlling the magnitude of response to various
cytokines including the interferons, gp130 cytokines, IL-12, IL-10, G-CSF, prolactin, growth hormone and IL-2 family cytokines. We have
recently shown SOCS4 to be a critical regulator of the anti-viral response, with loss of functional SOCS4 leading to increased
susceptibility to influenza infection (1). Given the similarity between the SOCS4 and 5 SH2 domains, and the lack of an obvious
phenotype in mice lacking SOCS5 protein (Socs5-/-), we have investigated its role during influenza infection. Socs5-/- mice were also
highly susceptible to infection with H1N1 Puerto Rico/8/34 (PR8) influenza A virus, displaying an increased morbidity and mortality,
which is associated with increased weight loss, elevated proinflammatory cytokines and delayed viral clearance. In contrast to Socs4deficient mice, these differences are evident at day 2 post-infection, suggesting that the innate immune response is perturbed. In
addition, Socs5 mRNA is expressed at significantly higher levels than Socs4 in lung epithelium, the primary site of viral replication, and
is upregulated in response to infection. An intrinsic defect in epithelial cells was confirmed by infection of cultured primary lung epithelial
cells from Socs5-/- mice. Moreover, wild-type bone marrow transfer into irradiated Socs5-/- mice did not rescue the phenotype,
confirming the non-hematopoietic nature of the defect. We are currently using proteomics and siRNA knock-down studies to identify the
innate signalling pathways that are regulated by SOCS5.
(1) Kedzierski et al., PLOS Pathogens 2014, 10(5):e1004134
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 96
279
Differential effects of pro-inflammatory cytokines IL-1β and TNFα on cell adhesion molecules in an in
vitro model of the human blood-brain barrier.
Dan Kho1, Vicky Nelson1, Lola Rotimi1, Simon O'Carroll1, Catherine Angel2
1. Centre for Brain Research, Auckland
2. School of Biological Sciences, University of Auckland, Auckland, New Zealand
The central nervous system (CNS) is an immune privileged site where highly specialised endothelial cells, which comprise the bloodbrain barrier (BBB), acts as a selectively permeable interface to control the passage of nutrients and inflammatory cells into the
brain. Disruption of the BBB and leukocyte infiltration are among the abnormalities seen in neuroinflammatory diseases such as
multiple sclerosis. Using a recently developed human cerebral microvascular endothelial cell line (hCMVEC), we investigated the effects
of two pro-inflammatory cytokines IL-1β and TNFα on the inflammatory response of these cells. Specifically, temporal expression of cell
adhesion molecules ICAM-1 (CD54) and VCAM-1 (CD106), and cytokine secretion were investigated using flow cytometry and multiplex
cytokine arrays. The endothelial cells demonstrated differential expression of cell adhesion molecules, where TNF-α induced higher
expression of ICAM-1 and VCAM-1 over the time course of 6 days. Quantification of soluble ICAM-1 and soluble VCAM-1 in endothelial
conditioned media revealed that more adhesion molecules were cleaved from the cell surface when cells were stimulated by IL-1β as
opposed to TNFα over a period of 72 hours. IL-1β induced a substantially different cytokine secretion profile in comparison to
TNFα. Our study shows that these two key pro-inflammatory cytokines differentially regulate the inflammatory response of brain
endothelial cells.
280
Effects of caffeic acid phenethyl ester on Prevotella intermedia lipopolysaccharide-induced
production of proinflammatory mediators in murine macrophages
Sung-Jo Kim1
1. Pusan National University School of Dentistry, Gyeongsangnam-do, South Korea, South Korea
Caffeic acid phenethyl ester (CAPE) has numerous potentially beneficial properties, including anti-oxidant, immunomodulatory and antiinflammatory activities. However, the effect of CAPE on periodontal disease has not been studied before. This study was designed to
investigate the efficacy of CAPE in ameliorating the production of proinflammatory mediators in macrophages activated by LPS
from Prevotella intermedia, a pathogen implicated in periodontal disease. CAPE exerted significant inhibitory effects
on P. intermedia LPS-induced production of NO, IL-1β and IL-6 as well as their mRNA expression in RAW264.7 cells. CAPE induced
HO-1 expression in cells activated with P. intermedia LPS, and selective inhibition of HO-1 activity by tin protoporphyrin IX significantly
attenuated the inhibitory effect of CAPE on LPS-induced NO production. CAPE did not interfere with IκB-α degradation induced
by P. intermedia LPS. Instead, CAPE decreased nuclear translocation and DNA binding activity of NF-κB p50 subunit induced with LPS.
In addition, CAPE showed strong inhibitory effects on LPS-induced STAT1 and STAT3 phosphorylation. Further, CAPE significantly
elevated the expression of SOCS1 mRNA in P. intermedia LPS-stimulated cells. Overall, this study suggests that CAPE
inhibits P. intermedia LPS-induced production of NO, IL-1β and IL-6 in murine macrophages through anti-inflammatory HO-1 induction
and inhibition of NF-κB, STAT1 and STAT3 activation, which is possibly related to the activation of SOCS1 signaling. Modulation of host
response by CAPE may represent an attractive strategy towards the treatment of periodontal disease. In vivo studies are underway to
further appraise the potential of CAPE as an immunomodulator in the treatment of periodontal disease.
This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded
by the Ministry of Education (2013R1A1A2007625).
281
Suppressors of Cytokine Signalling (SOCS) 1 and SOCS3 coordinate regulation of macrophage
polarization
Tatiana B Kolesnik1, Takashi Ushiki1, Edmond Linossi1, 2, Jian-Guo Zhang1, 2, Nikos A Nicola1, 2, Peter J Murray3, Warren S
Alexander1, 2, Sandra A Nicholson1, 2
1. The Walter & Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
2. Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
3. Department of Infectious Diseases and Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee,
USA
Macrophages are multi-functional innate immune cells, which play a critical role in the host defence against various pathogens such as
bacteria, viruses and parasites. Once activated, they produce cytokines and chemokines to promote cell recruitment and proliferation,
tissue repair and wound healing. They display a high degree of heterogeneity and phenotypic plasticity depending on their origin, the
detection of pathogen-associated molecular pattern molecules (PAMPs) and the surrounding cytokine milieu. At present macrophages
are broadly classified into M1 (classically activated) and M2 (alternatively activated), based on the activating stimuli and corresponding
expression of signature markers. The SOCS family proteins (CIS and SOCS1-7) play an important role in the negative regulation of
JAK-STAT signalling in various cytokine pathways. Genetic deletion of either Socs1 or Socs3 has a dramatic effect, resulting in
excessive cytokine-driven inflammation. In order to address the possible redundancy between SOCS1 and SOCS3 in regulating
macrophage polarization, we have generated CSF-1-cultured bone marrow-derived macrophages deficient in SOCS1, SOCS3 or both
SOCS1 and SOCS3, and polarized them in the presence of interferon (IFN)γ (M1), interleukin (IL)-4 (M2) or lipopolysaccharide (LPS).
Analysis of M1/M2 markers confirmed that SOCS1 is a negative regulator of IFNγ and IL-4 signalling, and that SOCS3 regulates
cytokine-driven STAT3 phosphorylation following LPS stimulation. Interestingly, compound deletion of SOCS1 and SOCS3
synergistically up-regulated both inducible nitric oxide synthase (iNOS) and arginase I (ArgI) in response to LPS, and ArgI in response
to IL-4 stimulation. In conclusion, we find no evidence for redundancy between SOCS1 and SOCS3 as each regulate specific cytokine
signalling cascades, however those signals converge on iNOS and ArgI. These two enzymes compete for a common substrate to
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 97
regulate production of nitric oxide, and thus SOCS1 and SOCS3 coordinate regulation of the iNOS/ArgI axis and an important antipathogen effector mechanism.
282
Cell crowding induces interferon regulatory factor 9 which confers resistance to chemotherapeutic
drugs
Iryna Kolosenko1, Katja Pokrovskaja1, Angelo De Milito1, Dan Grander1, Stig Linder1
1. Karolinska Institutet, Stockholm, Sweden
Background: Intrinsic tumor resistance to therapy remains a major problem in the management of cancer patients. Multiple mechanisms
are believed to contribute to this phenomenon. Recently, a subset of interferon-stimulated genes (referred to as interferon-related DNA
damage signature or IRDS) has been associated with poor therapy response and short overall survival for several cancer types. In this
study we aimed to investigate the mechanism of IRDS regulation in different cancer types.
Materials and methods: Using both 2D and 3D cancer cell line models, RNAi and over expression approaches, we investigated the
expression of IFN-stimulated genes using qRT-PCR and Western blotting approaches.
Results: a microarray analysis of a 2D and a 3D cultures( multicellular spheroids, MCS) of a human carcinoma cell line HCT116
revealed that IRDS genes are highly enriched in MCSs what reflects their relative resistance to chemotherapeutics. Analysis of a panel
of cancer cell lines (MCF7, SKOV3, DLD1, HCT116) demonstrated that IRF9 has a leading role in the regulation of interferon-stimulated
genes (e.g. STAT1, STAT2, IFITM1, IFI27, OAS1) and promoting therapy resistance. We have also demonstrated that this phenomenon
occurs in the absence of interferons and does not depend on STAT1, the main regulator of IFN-stimulated genes, but rather depends on
STAT2. Furthermore, we showed that a subset of IRDS is regulated by crowding and might depend on cell-to-cell contacts and/or
adhesion molecules.
Conclusion: Our findings demonstrate that IRF9 can act as a main regulator of interferon-stimulated genes rendering tumors resistant to
therapy. Therefore, it can potentially be developed as a marker of clinical response in oncology.
283
IL-1-Induced Production of Chemokines CCL5 and CXCL10 requires K63-linked Polyubiquitination of
IRF1
Kuzhuvelil B Harikumar1, Jessie W Yester1, Michael Surace1, Clement Oyeniran1, Megan M Price1, Wei-Ching Huang1, Nitai C
Hait1, Jeremy C Allegood1, Akimitsu Yamada1, Xiangqian Kong2, Helen M Lazear3, Reetika Bhardwaj1, Kazuaki Takabe1, Michael
S Diamond3, Cheng Luo2, Sheldon Milstien1, Sarah Spiegel1, Tomasz Kordula1
1. Virginia Commonwealth University, Richmond, Virginia, United States
2. Chinese Academy of Sciences, Shanghai, China
3. Washington University School of Medicine, St. Louis, Missouri, United States
IL-1 regulates a broad range of both immune and inflammatory responses and plays a critical role in autoinflammatory diseases.
Although IL-1 activates NF-kB that regulates many IL-1 controlled processes, IL-1 also induces expression of transcription factor IRF1,
yet the roles and mechanisms of its activation remain elusive. In contrast to TLRs that effectively activate IRF3 and IRF7 and induce
chemokine production and efficient recruitment of mononuclear cells to sites of infection, we show that the recruitment of mononuclear
cells to sites of sterile inflammation is coordinated by IL-1R-mediated activation of IRF1. We found that IRF1 is essential for IL-1induced expression of the chemokines CXCL10 and CCL5 that in turn recruit mononuclear cells into sites of sterile inflammation. Newly
synthesized IRF1 acquires K63-linked polyubiquitination mediated by cellular inhibitor of apoptosis 2 (cIAP2), whose E3 ligase activity is
enhanced by the bioactive lipid sphingosine-1 phosphate (S1P). In response to IL-1, cIAP2 and sphingosine kinase 1, the enzyme that
generates S1P, form a complex with IRF1, which leads to its activation. Thus, IL-1 triggers a novel signaling cascade that controls
induction of IRF1-dependent genes important for sterile inflammation. Targeting of this newly discovered IL-1-induced cascade may be
clinically important in the future.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 98
284
Identification of a novel low molecular weight inhibitor targeting macrophage migration inhibitory
factor (MIF).
Tali Lang1, Huapeng Fan1, Jacinta Lee1, James Harris1, Eric Morand1
1. Monash University Australia, Clayton, VIC, Australia
Aim: Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine involved in the regulation of both innate and adaptive immune
responses. As elevated levels of MIF are implicated in the pathogenesis of autoimmune disorders, it is an attractive target for
therapeutic intervention. Several classes of small-molecule inhibitors of MIF, including ISO-1, attenuate MIF’s pro-inflammatory
activities. We have identified a novel and potent MIF antagonist, COR123625, which is effective in vivo and in vitro.
Methods and Materials: To assess the effects of COR123625 in endotoxic shock in vivo, mice were treated with COR123625 (0.110mg/kg) by intraperitoneal injection 24 and 1 hour prior to administration of LPS (1.0mg/kg). Serum was collected after 90 min and
levels of TNF-α measured by ELISA. In vitro, murine immortalised bone marrow-derived macrophages (iBMMs) were pre-treated with
COR123625 or ISO-1 (0-100µM) prior to the addition of LPS (100ng/mL). ELISA was then used to measure levels of secreted cytokines
in cultured supernatants and qPCR to quantitate pro-inflammatory mRNA expression.
Results: At a concentration of 1 mg/kg, COR123625 significantly reduced LPS-induced serum TNF-α. This effect was not observed with
ISO-1 at doses between 0-20 mg/kg. Both COR123625 and ISO-1 significantly reduced LPS-induced expression of TNF-α and MKP-1
mRNA, while MCP-1 levels were unaffected. No inhibition of secreted TNF-α and MCP-1 were observed when compared to LPS only
cultured supernatants.
Conclusions: We show that COR123625 is a potent inhibitory compound against the promotion of TNF-α by MIF in vivo. In contrast to in
vivo findings, both COR123625 and ISO-1 were not as effective in reducing cytokine expression at the transcriptional and translational
level in vitro, which may be accounted for by cell-specific properties and suggests the effects observed in vivo may be dependent on
interactions with other cells.
285
Shedding light on preterm immunity
Chun Wang Jason Lao1, 2, Friederike Beker3, Kai König3, Elizabeth Noble3, Geraldine Walsh3, Atul Malhotra4, 2, Kenneth Tan4, 2,
Gregory Woodhead3, Claudia Nold-Petry1, 2, Marcel F Nold1, 2
1. Ritchie Centre, MIMR-PHI Institute of Medical Research, Clayton, VIC, Australia
2. Department of Paediatrics, Monash University, Clayton, VIC, Australia
3. Department of Paediatrics, Mercy Hospital for Women, Heidelberg, VIC, Australia
4. Monash Newborn, Monash Health, Melbourne, VIC, Australia
Background: There is little knowledge on the immune system of extremely premature infants. This paucity of evidence impedes
advances in the fight against bronchopulmonary dysplasia (BPD), a common, severe chronic lung disease that entails significant
morbidity and mortality. No safe and effective treatment exists.
Method: Blood was collected from extremely preterm infants at 5 timepoints [birth, days 1, 7 and 14 and 36 weeks corrected
gestational age (WCGA)]. Following overnight stimulation with PMA/ionomycin, LPS or vehicle in whole blood assays, flow cytometry
was used to explore T cells and their polarisation, macrophages and dendritic cells (DC) and their activation status and endothelial
progenitor cells (EPC).
Results: In preterm babies, expression of the activation marker MHC II progressively increased over time on macrophages (d1, 48% vs
36-WCGA, 86%) and DC (d1, 24% vs 36-WCGA, 56%). Comparing the one infant that developed BPD with the two that did not at 36WCGA, we observed a marked increase in macrophage (6-fold) and DC activation (2-fold), but fewer circulating EPC (0.3% vs 1.4% of
viable cells). Unexpectedly, we also found that preterm infants are capable of producing the cytokines IFN-gamma and IL-17A.
Conclusions: It only took the results from three preterm infants to reveal that this first-of-its-kind study will revolutionise the
understanding of preterm immunity; for example, these data suggest that, contrary to current belief, preterm babies are capable of
producing IFN-gamma and IL-17. BPD appears associated with markedly increased cellular activation - a promising finding that may
provide a basis for therapeutic innovations.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 99
286
Inhibitor of apoptosis (IAP) proteins repress spontaneous inflammatory joint disease
Kate Lawlor1, Mordechay Gerlic1, Cathrine Hall1, Sukhdeep Spall1, Holly Anderton1, Ian Wicks1, Warren Alexander1, W. WeiLynn (Lynn) Wong2, John Silke1, David Vaux1, James Vince1
1. Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
2. Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
Inhibitor of apoptosis proteins (IAPs), including x-linked (XIAP) and cellular IAP 1/2 (cIAP1/2), are important regulators of TNF receptor
induced cell death and cytokine production. We have previously demonstrated that, in vitro, IAP loss sensitises macrophages to TNF
induced caspase-8 activation and apoptosis, and limits RIPK3 mediated TNF and IL-1β secretion. Here we report that mice with genetic
deletion of XIAP and cIAP2 ubiquitously and cIAP1 in myeloid cells (c1LysMcrex-/- c2-/-) develop a mild spontaneous inflammatory arthritis
and dermal inflammation, which is associated with increased myelopoiesis and elevated cytokines, particularly IL-1β. In contrast, mice
doubly deficient in cIAPs (c1LysMcrec2-/-) but retaining XIAP, exhibit severe arthritis accompanied by elevated TNF levels. Predisposition to
inflammation upon IAP deletion in myeloid cells was further highlighted by increased disease severity in IL-1β dependent K/BxN serum
induced arthritis. Consistent with the role of IAPs as important negative regulators of RIPK3 and caspase-8 activity, we further show that
K/BxN arthritis is reduced in mice lacking RIPK3 alone, or in combination with apoptotic caspase-8. In contrast, deficiency in the
necroptotic effector MLKL did not alter disease pathogenesis. These results show that deletion of cIAP1 concurrently with either cIAP2
or XIAP is sufficient to cause spontaneous inflammation, albeit with distinct cytokine profiles. Furthermore, our findings overturn the
common assumption that reduced inflammatory disease in RIPK3-deficient mice is due to a block in necroptosis.
287
Role of STAT3 in regulation of cancer metabolism
David Levy1, Daniel Gough2, Isabelle Marié1, Camille Lobry1, Maher Abdul Hay1, Iannis Aifantis1
1. NYU School of Medicine, New York, NY, United States
2. MIMR-PHI Institute of Medical Research, Centre for Cancer Research, Clayton, Vic, Australia
STAT3 is a latent cytoplasmic transcription factor that responds to cytokine signaling and tyrosine kinase oncoproteins by nuclear
translocation when tyrosine phosphorylated and has been implicated in a wide variety of human cancers. We have found that malignant
transformation by activated Ras is impaired without STAT3, in spite of the inability of Ras to drive STAT3 tyrosine phosphorylation or
nuclear translocation. STAT3 mutants that cannot be tyrosine phosphorylated, are retained in the cytoplasm, or cannot bind DNA are
nonetheless capable of supporting Ras-mediated transformation. In addition to being a cytoplasmic protein that translocates to the
nucleus in response to tyrosine phosphorylation, STAT3 accumulates in mitochondria, and its presence in mitochondria is sufficient to
support Ras-mediated transformation. Mitochondrial STAT3 modulates metabolic output, influencing both glycolytic and oxidative
phosphorylation activities characteristic of cancer cells. These actions of mitochondrial STAT3 depend on its phosphorylation on serine
727, which is mediated by the MEK-ERK pathway in Ras-transformed cells.
K-Ras-dependent myeloid proliferative neoplasm in mice displays serine but not tyrosine phosphorylated STAT3. A point mutation
abrogating STAT3 S727 phosphorylation delayed onset and decreased disease severity in mice with oncogenic K-Ras expressed
in hematopoietic progenitors and significantly extended their survival. Activated K-Ras also required STAT3 for cytokine-independent
growth of myeloid progenitors in vitro, and mitochondrially restricted STAT3 and STAT3-Y705F, both transcriptionally inert mutants,
supported factor-independent cell growth. Although STAT3 was dispensable for growth of myeloid progenitors in response to cytokines,
abrogation of STAT3-S727 phosphorylation blocked K-Ras-driven, growth factor-independent malignant growth. These data document
that serine phosphorylated, mitochondrially-restricted STAT3 supports hematopoietic neoplastic cell growth induced by K-Ras. A series
of small molecules that impair growth of malignant cells appear to depend on the metabolic function of STAT3, suggesting that
mitochondrial STAT3 is a viable cancer target.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 100
288
Inhibition of JAK signalling by SOCS1
Nicholas PD Liau1
1. Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
Hypothesis
The Janus Kinase family of proteins (JAKs) are constitutively associated with the intracellular portion of cytokine receptors. Upon
cytokine stimulation of the receptor, JAKs provide the kinase activity necessary for the phosphorylation of substrate molecules, resulting
in a subsequent biological response. The Suppressors of Cytokine Signalling (SOCS) proteins inhibit JAK activity, regulating cytokine
signalling. SOCS1 and SOCS3 have a distinct method of inhibiting JAK1, JAK2 and TYK2 by competing with protein substrates, directly
inhibiting JAK catalytic activity.
We have solved the structure of SOCS3 in complex with JAK2 and a fragment of the gp130 receptor. This structure shows that SOCS3
binds JAK and receptor simultaneously. Importantly, SOCS3 is able to obtain specificity for the downregulation of certain cytokine
pathways by binding preferentially to some receptors.
In light of this information, we now aim to elucidate the mechanism through which the similar SOCS1 inhibits JAK catalytic activity.
Aims and methods
We have recently been able to recombinantly produce SOCS1, allowing structural and biophysical studies to be undertaken for the first
time.
Our preliminary data indicate that SOCS1 is a more potent inhibitor of JAK catalytic activity than SOCS3. We are currently investigating
the structural features of SOCS1 which contribute to such inhibitory activity. We are also investigating the features allowing SOCS1 to
specifically inhibit different cytokine pathways to those regulated by SOCS3.
Specifically, we aim to obtain the structure of SOCS1 bound to a JAK family member to understand its mode of inhibition. Further,
studies of engineered SOCS1 and JAK mutants can now reveal the direct contributions of particular regions of each protein to binding
and inhibition kinetics, as well as those regions which provide specificity of inhibition of certain cytokine pathways.
1.
2.
Kershaw, N.J., et al., SOCS3 binds specific receptor-JAK complexes to control cytokine signaling by direct kinase inhibition. Nature
structural & molecular biology, 2013. 20(4): p. 469-76.
Babon, J.J., et al., Suppression of cytokine signaling by SOCS3: characterization of the mode of inhibition and the basis of its
specificity. Immunity, 2012. 36(2): p. 239-50.
289
Understanding emergency haematopoiesis using cellular barcoding
Dawn Lin1, Shalin Naik1
1. WEHI, Melbourne, VIC, Australia
Haematopoiesis in the steady-state is reasonably well understood. However, upon emergency situations such as infection or
inflammation, homeostasis is broken and cell numbers can rise substantially e.g. neutrophil numbers after infection with Listeria
monocytogenes. What is not currently clear is whether this is due to the same progenitors producing more of a given cell type, or
instead if there is recruitment of 'emergency' progenitors for this purpose. Cellular barcoding is a novel technology that can track the
output of single stem and progenitor cells. Here, cells are transduced with lentivirus where each particle has a different DNA 'barcode'
that is integrated into the genome, and thus inherited by daughter cells. By establishing the barcode signatures in progeny cell types in
steady-state vs inflammatory conditions I aim to assess progenitor fate on the single cell level that gives rise to higher cell numbers. In
my model I will be assessing the affect of G-CSF and flt3 ligand on the output of several immune cell types.
1.
2.
Emergency granulopoiesis. Manz MG, Boettcher S. Nat Rev Immunol. 2014 May;14(5):302-14. Diverse and heritable lineage
imprinting of early haematopoietic progenitors.
Diverse and heritable lineage imprinting of early haematopoietic progenitors. Naik SH, Perié L, Swart E, Gerlach C, van Rooij N, de
Boer RJ, Schumacher TN. Nature. 2013 Apr 11;496(7444):229-32
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 101
290
Oncostatin M and IL-6 induce pro-inflammatory gene signatures in articular chondrocytes
Xiao Liu1, Ruijie Liu1, Ben A Croker2, Kate E Lawlor1, Gordon K Smyth1, Ian P Wicks1
1. Walter and Eliza Hall Institute of Medical Research, Melbourne, Vic, Australia
2. Boston Children's Hospital, Boston, Massachusetts, United States
Aims. This study describes gene expression in chondrocytes stimulated with two gp130 family cytokines - Oncostatin M (OSM) or IL-6 in
conjunction with soluble IL-6 receptor (IL-6/sIL-6R, i.e. IL-6 trans-signaling) and examines the impact of deleting Suppressor of Cytokine
Signaling-3 (SOCS3) in this cell type.
Methods. Wild type (WT) and SOCS3-deficient (Socs3Δ/Δcol2) murine primary chondrocytes were stimulated in vitro with OSM or IL-6/sIL6R, for 4 hours. Total RNA was extracted and expressed genes were identified by microarray analysis (Illumina MouseWG-6 v2.0
Expression BeadChip). Validation of microarray results was performed using Taqman probes on RNA derived from chondrocytes
stimulated with OSM or IL-6/sIL-6R for 1, 2, 4 or 8 hours. Gene set testing was undertaken using ROAST (rotation gene set testing).
Gene ontology was performed using DAVID v6.7.
Results. Preliminary data showed that chondrocytes rely on IL-6 trans-signaling. The gene transcription profiles between OSM and IL6/sIL-6R were highly correlated. Using pathway analysis, OSM was found to have more profound effects on chondrocyte gene
expression compared to IL-6/sIL-6R, and induced greater changes in biological processes, including “Cytokine activity”,
“Metallopeptidase activity” and “Proteolysis”. Further analysis distinguished between acute-phase (e.g. Ccl7) and late-stage
(e.g. Il19 and Saa1) changes in gene expression for both OSM and IL-6. In the absence of SOCS3, OSM and IL-6/sIL-6R stimulation
induced an IFN-like gene signature.
Conclusion. We found similarities and differences between OSM- and IL-6/sIL-6R-induced inflammatory gene signatures in
chondrocytes. SOCS3 plays an important regulatory role in this cell type, as it does in hematopoietic cells. Our results suggest OSM
may be a target for therapeutic intervention in inflammatory arthritis.
291
Targeting interleukin 6 in endothelial cells to prevent undesired formation of new blood vessels
Liza U Ljungberg1, Rongrong Wu1, Geena Paramel1, Veena Mishra1, Allan Sirsjö1
1. Div of Clinical Medicine, Dpt of Health and Medicine, Örebro University, Örebro, Sweden
Interleukin 6 (IL-6) is a multifunctional cytokine that plays a central role in inflammation by controlling differentiation, proliferation,
migration and apoptosis of targeted cells. In recent years it has been shown that IL-6 induces release of vascular endothelial growth
factor, and thus might play an important role in angiogenesis.
Aim: The present study aimed at investigating the involvement of IL-6 in angiogenesis by studying migration and tube formation of
vascular endothelial cells.
Methods: Migration and tube formation of human umbilical vein endothelial cells (HUVEC) were studied after treatment with IL-6, soluble
IL-6 receptor or the combination of IL-6 and the soluble IL-6 receptor. Migration and tube formation was also studied in HUVEC after
knock-down of IL-6 using siRNA. Knock down of IL-6 was confirmed using ELISA.
Results: Treatment with IL6 (50ng/ml), soluble IL-6 receptor (100 ng/ml) or the combination of IL6 and soluble IL6 receptor did not affect
tube formation or migration. Knock down of IL-6, on the other hand, resulted in reduced tube formation (number of tubes and total tube
length) as well as slightly reduced migration of HUVEC. This inhibition could be restored after addition of exogenous IL-6 (50ng/ml).
Conclusion: Our data suggests that IL-6 is involved in angiogenesis and that IL-6 produced by the endothelial cells themselves is
enough in to maintain normal tube formation and migration. Targeting IL-6 may be a novel strategy to prevent undesired formation of
new blood vessels.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 102
292
Detection, localisation and quantification of the interaction between IL-37 and its cell surface
receptor in human peripheral blood mononuclear cells via dSTORM super-resolution imaging
Camden Lo1, Kirstin Elgass2, Ina Rudloff1, 3, Claudia Nold1, 3, Marcel Nold1, 3
1. MIMR-PHI Institute, Clayton, VIC, Australia
2. Monash Micro Imaging, Monash University, Melbourne, Victoria, Australia
3. Department of Paediatrics, Monash University, Melbourne, Victoria, Australia
Aims
The elucidation of protein complexing and their cellular localisation are foundation to cytokine and signalling research. Traditional
methodologies such as immunoprecipitation and immunofluorescence cannot show complexing, localisation and abundance of
molecules simultaneously. We aim to co-opt super-resolution imaging techniques to study the interactions between the powerful antiinflammatory cytokine IL-37 and the receptor chains IL-18 receptor alpha (IL-18Ralpha) and SIGIRR. We describe cytokine-receptor
binding/complexing, subcellular localisation and molecular numbers of these three proteins in their endogenous, native context, i.e.
without exogenous additions, transfections or lysis.
Methods
The basis for our technique is direct stochastic optical reconstruction microscopy (dSTORM, 1). AlexaFluor488-, AlexaFluor568- and
AlexaFluor647-tagged secondary antibodies were used to label the ligand (IL-37) and the two subunits of its heterodimeric receptor
complex (SIGIRR and IL-18Ralpha) in PBMC from healthy human donors. The fluorophores were induced into a dark state by high
intensity laser illumination and reducing buffer conditions. The stochastic return of the fluorophores to an active state is imaged and
statistically analysed, allowing detection and localisation at 20-50nm precision. We then derive absolute localisation (positioning) and
proximity of all three proteins relative to each other on the cell surface, and infer complexing and interaction based on clustering and colocation at molecular binding distances. As the technique is single molecule sensitive, we further measure and sample the each protein
at the cell surface and derive the molecular proportion that is involved in complexing.
Results and conclusions
IL-37 forms a ligand:receptor complex with SIGIRR and IL-18Ralpha (sub 50nm proximity with each other) on the cell surface of
primary, untransfected human PBMC. Thirty min after LPS, approximately 15% of SIGIRR and 6% of IL-18Ralpha molecules are
involved in complexing with IL-37 on the cell surface. This is the first demonstration of any super-resolution technique to study three-way
protein interactions in primary human samples.
1.
1. Super-resolution imaging with small organic fluorophores. Heilemann M, van de Linde S, Mukherjee A, Sauer M. Angew Chem
Int Ed Engl. 2009;48(37):6903-8. doi: 10.1002/anie.200902073.
293
Regulation of myeloid cell population kinetics and phenotype by CSF-1 and GM-CSF
Cynthia Louis1, Andrew Cook1, Derek Lacey1, John Hamilton1
1. University of Melbourne, Parkville, VIC, Australia
GM-CSF (CSF-2) and CSF-1 are key cytokines for the mononuclear phagocytes, including monocytes, macrophages and dendritic cells
(DCs). GM-CSF is critical for in vitro monocyte-derived DCs (Mo-DCs) generation and the development of resident CD103+ DCs in
some tissues. In contrast, CSF-1 is essential for the homeostasis of monocyte/macrophage lineage cells. However, clear delineation of
DC subsets, Mo-DCs and classical DCs (cDCs), has remained relatively unclear because of many overlapping features of these cells
such as surface markers, making it difficult to discern the differential roles of GM-CSF or CSF-1 in controlling such cells.
In this study, we assess the effects of GM-CSF and CSF-1 on myeloid-lineage populations during inflammation utilizing a number of
animal models. Peritoneal exudate cells (PECs) were taken from antigen (mBSA)-induced peritonitis (AIP) or thioglycolate-induced
peritonitis. Spleen cells from intravenous LPS-challenged mice were examined. Mice also received anti-GM-CSF (22E9), anti-M-CSFR
(AFS98), or control mAbs. Monocytes, macrophages, and DC subsets (Mo-DCs and cDCs) were monitored by flow cytometry.
We classified all monocyte descendants, including monocytes, monocyte-derived macrophages and Mo-DCs, by their CSF-1
receptor/CD115 expression, setting them apart from cDC lineage. Inflammatory Mo-DCs were found in the AIP PECs, but not in
thioglycolate-PECs or LPS-challenged spleens, and were depleted specifically in GM-CSF-/- or anti-GM-CSF-treated mice. AIP Mo-DCs,
macrophages, and monocytes were all depleted following CSF-1R neutralization. In contrast, cDCs subsets in all tissues examined
were relatively unaffected by the GM-CSF or CSF-1R neutralization. In summary, GM-CSF deletion or anti-GM-CSF treatment
abrogated the accumulation of inflammatory Mo-DCs, with no effect on nonlymphoid tissue cDCs, in T cell-dependent inflammation; in
contrast, the CSF-1 receptor was important for all inflammatory monocyte descendants, but not the cDC lineage populations.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 103
294
Understanding the Role of Pro-inflammatory Cytokines in the Development of Gastric Cancer
Jun Ting Low1, Tracy Putoczki1, Andreas Strasser1, Lorraine A O'Reilly1
1. The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
NF-κB, in particular the RelA subunit, is an important transcriptional regulator of many genes involved in tumour-promoting
inflammation, cell proliferation and survival, and has frequently been found to be aberrantly over-activated in infiltrating immune cells in
gastritis and in gastric cancer (GC). Our laboratory has discovered that ageing NF-κB1-deficient (nfκb1-/-) mice develop GC in a manner
that mimics the progression of human invasive intestinal-type GC. These mice develop chronic gastritis, gastric mucosal atrophy or
dysplasia, with a high incidence of invasive gastric adenocarcinomas. We observed abnormally increased infiltration of F4/80-positive
macrophages and CD45-positive leukocytes in the stomachs of young nfκb1-/- mice. Concurrent with the recruitment of inflammatory
cells into the gastric mucosa, elevated levels of pro-inflammatory cytokines and chemokines were found in the serum of young nfκb1-/mice, and also in established tumours of these mice. This implicates deregulated expression of pro-inflammatory cytokines in GC
disease progression.
Inflammatory cells recruited into the gastric mucosa during chronic gastritis secrete pro-inflammatory cytokines and chemokines.
Interestingly, loss of NF-κB1 has been found to exacerbate macrophage M1-driven inflammation, augmenting pro-inflammatory cytokine
production. In order to characterise the role of abnormally elevated cytokines in the onset and progression of GC in nfκb1-/- mice, we
generated compound mutant mice lacking both NF-κB1 and individual pro-inflammatory cytokines that have been linked to cancer
development. The survival, histopathology, immune cell infiltration, cytokine and chemokine levels of these mice is being examined at
multiple time points. We have also characterised the cell populations in the gastric mucosa and have quantified the levels of
phosphorylated-Stat3 and phosphorylated-p65/NF-κB levels in gastric epithelial cells and infiltrating leukocytes of these compound
mutant mice. Our results determine the roles these cytokines have in driving GC in the nfκb1-/- mice and highlight the therapeutic
potential in targeting pro-inflammatory cytokines for the treatment of GC.
295
Therapeutic deletion of IL-17 producing CD8+ T cells (Tc17) attenuates GVHD without impairing GVL.
Kate H Gartlan1, kate A Markey1, Mark Bunting1, antiopi varelias1, glen boyle1, Steven Lane1, Motoko Koyama1, Geoff Hill1, Kelli
PA MacDonald1
1. QIMR, Brisbane, QLD, Australia
Donor T cell polarization is a critical factor influencing the severity and tissue distribution of graft-versus-host disease (GVHD) and the
potency of graft-versus-leukemia (GVL) effects after bone marrow transplantation. We have recently reported that G-CSF treatment
promotes type-17 differentiation in both CD4 and CD8 T cells and that donor IL-17A, predominantly from CD8 T cells (Tc17), mediates
fibrotic skin pathology manifesting late after transplant as scleroderma. To study Tc17 development and function we utilized the IL17ACreRosa26ReYFP ‘fate-mapping’ reporter mouse and observed that donor Tc17 cells differentiate early post allogeneic transplant
and transition rapidly towards a Tc1-like phenotype. Tc17 differentiation is dependent upon IL-6, host-DC and is regulated by the
presence of IFNγ. Tc17 cells appear highly inflammatory, displaying considerable promiscuity in their transcriptional profile and
inflammatory cytokine production. Furthermore, targeted deletion of Tc17 early post transplant was protective in a lethal model of acute
GVHD. In contrast, Tc17 express only low levels of the CTL effector molecule Granzyme B and display strikingly poor GVL activity in
vivo. These data demonstrate that Tc17 differentiation is an early and highly plastic differentiation program, culminating in a poorlycytolytic, inflammatory population that mediates GVHD without contributing to GVL. Thus, early therapeutic targeting of Tc17
development via IL-6 inhibition represents a highly attractive avenue for GVHD prevention.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 104
296
PB1-F2 from H7N9 Influenza A virus activates the NLRP3 inflammasome to induce inflammation
Anita Pinar1, Michelle Tate1, Julie McAuley2, Jen Dowling1, Eicke Latz3, 4, Lori Brown2, Avril Robertson5, Matthew
Cooper5, Ashley Mansell1
1. Centre for Innate Immunity and Infectious Diseases, MIMR-PHI Institute of Medical Research, Monash University, VIC, Australia
2. Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC, Australia
3. Institute of Innate Immunity, University of Bonn, Bonn, Germany
4. Division of Infectious Diseases & Immunology, University of Massachusetts Medical School, Worcester, MA, USA
5. Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
Emerging influenza A virus (IAV) strains that cross over into humans present a global health danger due to their potential high mortality.
These pandemic infections are characterized by the hyper-inflammatory response, or ‘cytokine storm’ which can lead to severe clinical
symptoms and death.
We recently identified the virulence factor PB1-F2 in the highly pathogenic mouse-adapted A/PR8 IAV strain as a potent activator of the
NLRP3 inflammasome, inducing secretion of mature pyrogenic IL-1b. Activation of the inflammasome was directly linked to pathogenic
IAV aetiology. We have now identified that PB1-F2 peptides derived from highly pathogenic strains of the 20 th century are also able to
induce IL-1b secretion.
Recent sporadic infections of humans in China with avian H7N9 IAV have caused concern due to high mortality, yet virulence factors
associated with severe pathology are unclear. H7N9 viruses isolated from human express full length PB1-F2 which may act as a
virulence factor. We have found that PB1-F2 peptide derived from H7N9 is a potent inducer of IL-1b secretion, comparable to A/PR8.
Importantly, IL-1b secretion is reduced by inhibiting phagocytosis, lysosomal acidification and Caspase-1 activation while macrophages
deficient in ASC, Caspase-1 and NLRP3 display ablated IL-1b secretion. Our data therefore identify H7N9 PB1-F2 as a novel
inflammasome activator. Critically treatment of mice with H7N9 PB1-F2 peptide induced leukocyte infiltration and IL-1b secretion into
the lung airspaces.
Given the high mortality associated with pathogenic IAV outbreaks, we further investigated if suppression of the inflammasome with the
inhibitor MCC950 could reduce inflammation. We have subsequently found that MCC950 is able to potently inhibit PB1-F2-induced IL1b secretion and inflammasome activation. Importantly within the context of pathogenic IAV outbreaks, treatment of mice with MMC950
three days post-intranasal infection with pathogenic PR8 IAV delays the onset of disease.
These findings suggest that H7N9 IAV pathogenicity may be associated with PB1-F2-induced inflammasome activation and that
targeted inhibition of the inflammasome may provide a therapeutic intervention to alleviate the excessive inflammation associated with
pathogenic IAV infections.
297
Positive and Negative Epigenetic Regulatory Steps During IFN-Stimulated Transcriptional Initiation
and Elongation
Isabelle Marié1, Hao-Ming Chang1, Leonid Gnatovskiy1, David Levy1
1. NYU School of Medicine, New York, NY, United States
IFN-stimulated gene (ISG) expression is mediated by the ISGF3 transcription factor complex, composed of tyrosine phosphorylated
STAT1 and STAT2 and the DNA binding partner, IRF9. The transactivation domain of STAT2 recruits transcriptional activators and
chromatin modifiers that regulate polymerase recruitment and elongation. Interestingly, both histone acetyltransferase (HAT) and
histone deacetylase (HDAC) activities are required for transcription of ISGs and establishment of an antiviral state. Inhibition of HDAC
activity or reduction of HDAC1, 2, and 3 abrogates ISG transcription without altering the activation or chromatin recruitment of ISGF3.
To pinpoint this HDAC requirement, we examined ISG transcription in vitro and ISG epigenetic regulation in vivo. While transcription of
ISGs on nucleosome-free DNA in vitro was unaffected by HDAC activity, chromosomal remodeling occurred at ISG promoters in vivo in
response to IFN stimulation and this remodeling required HDAC activity. To discover factors required for ISG transcription, we purified
native ISGF3 complexes and identified STAT2-interacting proteins by mass spectrometry. The DNA helicases Rvb1 and Rvb2
associated with the transactivation domain of STAT2 and reducing their expression by RNA interference impaired ISG transcription.
Neither Rvb1 nor Rvb2 were required for induction of IFN-gamma or TNF-alpha induced transcription. Interestingly, IFN-alpha
stimulation recruited RNA Pol II to ISG promoters even in the absence of HDAC activity, and the recruited Pol II became phosphorylated
on Ser-5, a hallmark of transcriptional initiation. However, RNA Pol II did not become phosphorylated on Ser-2 and failed to transcribe
IFN target genes in the absence of HDAC activity. Moreover, BRD proteins, PAF1, and SAGA complexes were found to regulate ISG
transcription, including requirements for regulated and possibly sequential histone acetylation, deacetylation, and ubiquitinylation.
Individual chromatin modification steps were impaired when HAT or HDAC enzymes were inhibited. These data define an ordered set of
chromatin modifications that coordinate individual regulatory events necessary for recruitment of positive factors and dismissal of
negative factors during the ISG transcriptional initiation and elongation cycle.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 105
298
Investigating type I interferon gene signatures in breast cancer patients
Zoe Marks1, 2, Helen Cumming1, 2, Jodee Gould1, 2, Alamgeer Muhammad2, Belinda Parker3, 2, Vinod Ganju2, Paul Hertzog1, 2
1. MIMR-PHI Institute, Clayton, VIC, Australia
2. Monash University, Clayton, VIC, Australia
3. La Trobe University, Melbourne, VIC, Australia
Bone metastasis is a key characteristic of end-stage breast cancer and severely reduces patient survival. While staging and grading of
primary tumour cells help to characterise the invasive nature of the tumour, currently there is no way of definitely predicting prognosis in
breast cancer patients. Interferon regulatory factor 7 (IRF7) is a transcription factor known to be a key regulator of type I IFN. We
recently showed that the loss of IRF7 expression in bone metastases compared to primary breast tumour cells results in reduced type I
IFN activation of peripheral anti-tumour immunity; thus enabling metastasis 1. The aim of this project is to investigate the type I IFNregulated gene expression in human peripheral blood and whether a signature exists in breast cancer patients as a potential marker for
prognosis. Transcription profiling microarray experiments will be performed using peripheral blood mononuclear cells (PBMC) from
healthy donors; breast cancer patients who responded to chemotherapy; and non-responders. The gene signature derived in the
previous aim will be refined using Interferome v2.02 and 'Enrich' and used to examine whether the type I IFN-regulated gene signature is
expressed differentially as a reflection of the presence or absence of an intact IFN/IRF7 pathway in primary breast cancer cells and
further, whether differential expression indicates prognosis.
1.
2.
Bidwell BN, Slaney CY, Withana NP, Forster S, Cao Y, Loi S, et al. Silencing of Irf7 pathways in breast cancer cells promotes bone
metastasis through immune escape. Nat Med. 2012;18(8):1224-31.
Rusinova I, Forster S, Yu S, Kannan A, Masse M, Cumming H, et al. Interferome v2.0: an updated database of annotated
interferon-regulated genes. Nucleic Acids Res. 2013;41(Database issue):D1040-6.
299
Arid5a enhances the development of Th17 cells by stabilizing Stat3 mRNA
Kazuya Masuda1, Barry Ripley1, Kishan Kumar Nyati1, Praveen Kumar Dubey1, Mohammad Mahabub-Uz Zaman1, Hamza
Hanieh1, Tadamitsu Kishimoto1
1. Immune regulation, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
Th17 cells play a critical role not only in host defense and mucosal defense but also in tissue inflammation and autoimmunity. Although
the mechanism of regulation of Th17 cell differentiation at transcriptional level is extensively studied, little is known about the posttranscriptional gene regulation in Th17 cells. Here we report for the first time that AT-rich interactive domain 5a (Arid5a) controls the
differentiation of Th17 cells through stabilization of Stat3 mRNA. Arid5a is specifically induced under Th17 cell polarizing condition but
not Th1, Th2, and Treg cell condition. Consequently, enhanced Arid5a protein binds to the stem-loop sequence of the 3’ untranslated
region (UTR) of the Stat3 mRNA, in turn, stabilizes Stat3 mRNA by competing with Regnase-1 on the same portion. Conversely, Arid5a
deficiency led to reduction of the frequency of Th17 cell population, in which the expressions of the Stat3-regulating gene, Rorc, IL21, IL-23R was significantly lower than those of WT T cells. Moreover, we have shown that such intrinsic role of Arid5a in T cells is
critical for induction of experimental autoimmune encephalomyelitis (EAE) in Rag2 deficient mice by adoptively transferred T cells. Thus,
we demonstrated that Arid5a plays an important role in the differentiation of Th17 cells through control of stabilization of Stat3 mRNA,
which results in exacerbation of autoimmune inflammation.
ILK inhibition. Instead, ILK is involved in an alternative activation of NF-kB signaling by modulating the phosphorylation of p65 at
Ser536. Furthermore, ILK-mediated alternative NF-kB activation through p65 Ser536 phosphorylation also occurs during Helicobacter
pylori infection in macrophages and gastric cancer cells. Moreover, ILK is required for H. pylori-induced TNF-α secretion in
macrophages. While ILK-mediated phosphorylation of p65 at Ser536 is independent of the phosphotidylinositol 3-kinase (PI3K)/Akt
pathway during LPS stimulation, upon H. pyloriinfection this event is dependent on the PI3K/Akt pathway. Moreover, in mouse model of
DSS-induced colitis, DSS-induced colon tissue damages, macrophages infiltration and colitis disease activity (measured by colon
length, body weight loss and disease activity index) were significantly reduced in ILKfl/fl;LysMCre mice compared to the control mice,
indicating a critical role of ILK in inflammatory diseases.
Conclusions:
Our findings implicate ILK as a critical regulatory molecule for pro-inflammatory signaling and a potential target for therapeutic
intervention in inflammatory diseases.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 106
300
Decidual-trophoblast interactions are critical for maternal tolerance of pregnancy.
Ellen Menkhorst1, Amy Winship1, Natalie Lane1, Joanne Yap1, Evdokia Dimitriadis1
1. MIMR-PHI Institute, Clayton, VIC, Australia
Invasion of specialized fetal cells, the ‘extravillous trophoblast’ (EVT) into the uterine decidua is critical for the establishment of
pregnancy. This presents an immunological paradox – why do invasive EVTs escape rejection?
One hypothesis is that decidual leukocytes are ‘specialized’ compared to peripheral leukocytes with pregnancy-specific functions.
However, the mechanisms leading to the differentiation of decidual-specific leukocyte populations are not well understood. The local
endometrial environment likely regulates leukocyte differentiation and activation. Prior to implantation endometrial stromal cells (ESC)
differentiate to become ‘decidualized’ and decidual leukocyte numbers increase such that during the 1st trimester, the period of maximal
EVT invasion, around 30-40% of cells in the endometrium are leukocytes, predominantly uterine Natural Killer cells and macrophages.
Abberant leukocyte activation and impaired decidualization are features of recurrent miscarriage and preeclampsia. We aimed to
investigate how interactions between decidual cells and the invading EVTs might influence maternal tolerance.
Cytokine secretion by primary human decidual biopsies (n=3) was identified by Luminex. Primary human ESC were decidualized in vitro
and conditioned media (CM) collected from non-decidualized and decidualized ESC. Primary human EVT (n=6) were treated with
pooled (n=14) ESC CM and secreted proteins <30kD identified by mass spectrometry and validated by immunohistochemistry and
Western blot.
Decidual biopsies secreted high levels of G-CSF, GM-CSF, IL6, IL8 and MCP1. The presence of EVTs within this tissue elevated IL1RA
secretion and reduced IL6, IL15 and MCP1 secretion. EVTs exposed to decidualized CM expressed cell surface factor CD59, which
inhibits formation of the complement membrane attack complex. In contrast, EVTs exposed to non-decidualized CM secreted proinflammatory proteases including Annexin A2, which can activate macrophages via TLR4 and DPP1, which activates granzymes.
Overall, our data suggests decidualization and appropriate interactions between ESC and EVTs are critical for maternal tolerance.
Impaired decidualization may lead to EVT expression of factors which aberrantly activate maternal leukocytes.
301
Mitochondrial apoptosis is dispensable for NLRP3 activation but non-apoptotic caspase-8 is required
for inflammasome priming
Ramanjaneyulu Allam, Kate Lawlor1, 2, Chi Wang Yu3, Donia M Moujalled1, 2, Rowena S Lewis1, 2, Francine Ke1, 2, Alison
Mildenhall1, 2, Kylie D Mason1, 2, Lorraine A O’Reilly1, 2, Michael J White1, 2, Andreas Strasser1, 2, David L Vaux1, 2, John Silke1, 2,
Warren Alexander1, 2, Benjamin T Kile1, 2, James E Vince1, 2
1. Walter and Eliza Hall Institute, Parkville, VIC, Australia
2. Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
3. Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
The NOD-like receptor protein 3 (NLRP3) inflammasome senses a variety of pathogen, host and environmental molecular patterns to
mediate caspase-1 activation, thereby promoting caspase-1 processing and secretion of the pro-inflammatory cytokines IL-1b and IL-18.
The current paradigm states that mitochondrial damage is a critical determinant of NLRP3 inflammasome activation. Here, we
genetically assess whether mitochondrial signalling represents a unified mechanism to explain how NLRP3 is activated by divergent
stimuli. Neither co-deletion of the essential executioners of mitochondrial apoptosis BAK and BAX, nor removal of the mitochondrial
permeability transition pore component cyclophilin D, nor loss of the mitophagy regulator Parkin, nor deficiency in MAVS affects NLRP3
inflammasome function. In contrast, caspase-8, a caspase essential for death-receptor-mediated apoptosis, is required for efficient Tolllike-receptor-induced inflammasome priming and cytokine production. Collectively, these results demonstrate that mitochondrial
apoptosis is not required for NLRP3 activation, and highlight an important non-apoptotic role for caspase-8 in regulating inflammasome
activation and pro-inflammatory cytokine levels.
302
Clostridium difficile toxin B induces an inflammatory cytokine response in a mouse model of
infection
Steven Mileto1, Anjana Chakravorty1, Glen Carter1, Dena Lyras1
1. Monash University, Clayton, Vic, Australia
Clostridium difficile is an important nosocomial pathogen of humans. Currently the role of toxin A and toxin B in disease pathogenesis
and in the development of severe disease resulting from C. difficile infection (CDI) is poorly characterised. Furthermore, the factors that
predispose some patientsto develop mild disease while others develop life-threatening disease are unknown. Understanding how C.
difficile modulates the host immune response during infection may provide insight into this phenomenon and may provide a better
understanding of the role that each toxin plays in disease. In this study, we examined the role of both toxins in pathogenesis and the
induction of the host immune response with disease progression. The analysis of an isogenic panel of independently derived toxin gene
mutants in a BI/NAP1/027 C. difficile strain using the mouse model of CDI showed that toxin B plays a critical role in the development of
severe disease and in the induction of the host immune response. Increased levels of pro-inflammatory cytokines and chemokines such
as C5a, TNF-α, MCP-1 and CXCL1, 9 and 10 were found in colonic tissue isolated from mice infected with a toxin A mutant, at levels
similar to those detected in tissues from mice infected with the wild-type strain. Cytokine induction coincided with the development of
severe colonic damage and fulminant disease. By contrast, infection with toxin B mutants resulted in only self-limiting diarrhoea and mild
disease and led to the induction of significantly lower levels of pro-inflammatory cytokines than observed in mice infected with the wildtype strain. Toxin B therefore appears to be the major virulence factor of C. difficile with toxin A playing a more minor role. This study
provides valuable insights into the role of toxin A and toxin B in C. difficile disease and the host immune response
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 107
303
Cytokines differentially regulate complement receptor immunoglobulin (CRIg) in human
macrophages: a control point in inflammation
Usma Munawara1, 2, 3, Kanchana Usuwanthim1, 2, 4, Yuefang Ma1, Alex Quach1, 2, Nick Gorgani1, 2, Charles Hii1, 2, Catherine
Abbott3, Antonio Ferrante1, 2, 4, 5, 6
1. Department of Immunopathology, SA Pathology at Women’s and Children’s Hospital, North Adelaide, South Australia
2. School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, Adelaide
3. School of Biological Sciences, Flinders University; Bedford Park, South Australia
4. Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Thailand
5. Discipline of Microbiology and Immunology, University of Adelaide , South Australia,
6. School of Pharmacy and Medical Sciences, University of South Australia., South Australia.
Aim(s)
and
Introduction
CRIg, a recently characterized complement receptor, is structurally and functionally distinct from the classical complement receptors,
CR3/CR4. CRIg expression is restricted to macrophages. While these receptors promote phagocytosis, CRIg also expresses antiinflammatory/immunosuppressive activity. Reduced ratio of CRIg to CR3 expression in macrophages skews the response towards
inflammation. We propose that CRIg is a control point in inflammation and its expression is regulated by cytokines in a manner that is
commensurate with the action of the cytokines (that cytokine actions occur via regulating CRIg expression).
Methods
Cytokines were examined for their effects on (i) the differentiation of human monocytes to CRIg+ macrophages in culture, (ii) CRIg
expression in monocyte-derived macrophages (MDM) and (iii) CRIg expression in dendritic cells (DC) generated from GM-CSF/IL-4
treated monocytes. The range of cytokines investigated included Th1, Th2, pyrogenic, regulatory and haematopoietic growth factors
types (LT, IFN-γ, IL-4, IL-13, TNF-α, IL-1β, IL-6, IL-10, TGF-β1, M-CSF and GM-CSF). CRIg mRNA was measured by qRT-PCR and
protein by Western blot and/or flow cytometry.
Results
Development of CRIg+ macrophages and CRIg expression in MDM and DC was positively or negatively regulated by cytokines. The
changes caused by these cytokines were dependent on the macrophage type or process and conducive with their role in promoting or
protecting against inflammation associated with rheumatoid arthritis and atherosclerosis. The cytokines similarly regulated both spliced
forms of CRIg. The results also showed that the cytokines differentially regulated CR3/CR4 compared with CRIg expression.
Furthermore, using PKCα-deficient MDM generated by RNAi, our data demonstrated that PKCα plays an important role in regulating
CRIg expression in macrophages and that cytokines such as TNF-α regulated CRIg expression via PKCα.
Conclusions
Cytokines regulate the development of CRIg+ macrophages and CRIg expression on macrophages and DC, in a manner conducive with
their role in inflammation.
304
Candida induces pyroptosis during escape from macrophages
Thomas Naderer1
1. Monash University, Clayton, VIC, Australia
The human fungal pathogen Candida albicans exists as commensal yeast cell but becomes invasive as hyphae. While several pattern
recognition receptors recognize the carbohydrate rich cell wall of Candida, yeast and hyphae induce different cytokine and immune
responses. This is largely because intracellular hyphae, but not yeast cells, are detected by the NLRP3 inflammasome leading to
caspase-1 activation and Il-1β secretion. Here we show activation of the NLRP3 inflammasome by intracellular Candida also induces
pyroptosis. In the absence of NLRP3, ASC or caspase-1, Candida is still able to form intracellular hyphae but fails to kill and escape
from macrophages soon after phagocytosis. Conversely, we have generated Candida mutants that still form hyphae but display reduced
levels of caspase-1 activation, pyroptosis and Il-1β secretion. These mutants show a breakdown of the cell wall architecture, in
particular reduced expression of glucans, which are known targets of macrophage receptors. Importantly, despite forming hyphae in
susceptible mice these Candida mutants are heavily attenuated in virulence, suggesting that pyroptosis is a critical escape mechanism
of Candida. This is surprising, as we have recently shown that intracellular hyphae can also induce delayed macrophage death to
escape, even in the absence of pyroptosis and other programmed cell death pathways. Thus, we provide the first report of a fungal
pathogen to induce pyroptosis in macrophages and provide evidence that early macrophage escape by hijacking pyroptosis contributes
toCandida virulence.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 108
305
Inhibition of IFN-regulated mediators of the IRF3 signaling pathway using small molecule inhibitors
targeting TBK1
Devi Ngo1, Louisa Phillipson1, David Segal1, Ian Wicks1, Chris Burns1
1. walter and eliza hall institute of medical research, Parkville, VIC, Australia
Aims
Interferon-regulatory factor 3 (IRF3) is a key transcription factor that mediates TLR3 and TLR4 activation of IFN-regulated gene
expression in inflammatory diseases such as rheumatoid arthritis (RA). The IKK-related kinase, TANK-binding kinase 1 (TBK1), is
essential for IRF3 activation. Here, we investigate the in vitro effects of a novel small molecule inhibitor that selectively inhibits TBK1
and inflammatory mediators downstream of IRF3 signaling.
Methods
TBK1 kinase assays were performed using recombinant TBK1 protein in an ADP luminescent kinase assay. The ability of TBK1
inhibitors (WEHI-112 and MRT67307) to inhibit LPS induced phosphorylation of IRF-3 was assessed in primary mouse synoviocytes
and human THP-1 macrophages by Western Blot. The effect of TBK1 inhibitors on LPS or Poly I:C induced cytokine production (IP10,
RANTES and TNF) by RAW264.7 cells was measured in cell culture supernatant by ELISA. IFNα/β gene expression and protein levels
in culture supernatants were detected using QPCR and the B16-reporter cell line, respectively.
Results
The TBK1 inhibitor WEHI-112 suppressed TBK1 activity with similar potency to the previously described compound MRT67307
(IC50 14nM and 43nM respectively). Western blot analysis demonstrated that WEHI-112 inhibited LPS and Poly I:C-induced
phosphorylation of IRF3 in mouse synoviocytes and THP-1 macrophages, comparable to MRT67307. In addition, both WEHI-112 and
MRT67307 potently inhibited LPS- and poly I:C-induced IP10, RANTES and type I IFN production by RAW macrophages. TBK1
inhibitors failed to modulate TNF production, demonstrating selectivity for the IRF3 signaling pathway.
Conclusion
We have developed a novel, small molecule inhibitor of TBK1 that potently suppresses TBK1 activated IRF3 phosphorylation and the
production of IP10, RANTES and type I IFN. This study demonstrates a promising avenue for the development of small molecule
inhibitors targeting TBK1 as a potential therapeutic in inflammatory diseases, including RA.
306
Understanding the Role of Cytokines in the Onset and Progression of Intestinal-Type Gastric Cancer
Paul Nguyen1, 2, Rita Busuttil3, Lisa Mielke1, 2, Gabrielle Belz1, 2, Alex Boussioutas3, Matthias Ernst1, 2, Tracy Putoczki1, 2
1. Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
2. Department of Medical Biology, Melbourne University, Parkville, VIC, Australia
3. Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
Gastric cancer (GC) is the fourth most prevalent cancer worldwide, and the third most common cause of cancer-related death. The
disease is generally asymptomatic and as a result is often diagnosed at an advanced stage, at which point metastasis is present in
greater than 80% of patients. Treatment options for GC are limited; however, current therapeutic strategies have embraced the concept
of targeting components of the inflammatory microenvironment. In light of this, we aimed to characterise the role of cytokines produced
by mucosal immune cell populations during onset and progression of intestinal-type gastric cancer (IGC). We compared microarray data
from a panel of >40 human IGC biopsies and adjacent non-tumour tissue to characterise the expression of cytokines and transcription
factors classically associated with chronic inflammation. Since we could detect numerous pro-inflammatory cytokines in human IGC
tumour tissue, we characterised the role of individual cytokines in disease progression using a validated mouse model of IGC, referred
to as gp130Y757F (1). We found that genetic restriction of IL-10 and IL-12 (p40) in did not alter disease progression in gp130Y757F mice.
Meanwhile, loss of IL-6, IL-17A and IL-23 (p19) expression delayed gastritis, but did not alter the progression of established gastric
adenomas. In contrast, genetic inhibition of IL-18, or therapeutic inhibition of IL-22 significantly reduced gastric tumour burden. Our
results suggest stage-specific roles for various cytokines during IGC progression, and identify IL-18 and IL-22 as potential therapeutic
targets for this disease.
1.
Cancer Cell. (2013). 24(2): 257
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 109
307
A Two-Site Interaction Underpins TRIM25 Activation of the RIG-I Anti-Viral Response
Akshay D'Cruz1, 2, Nadia Kershaw1, 2, Edmond Linossi1, 2, Laura Dagley1, 2, Jessica Chiang3, May Wang3, Thomas Hayman1, 2,
Jian-Guo Zhang1, 2, Michaela Gack3, Nicos Nicola1, 2, Jeffrey Babon1, 2, Sandra Nicholson1, 2
1. Inflammation Division, Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
2. The University of Melbourne, Parkville, VIC, Australia
3. Department of Microbiology & Immunobiology, Harvard Medical School, Boston, MA, United States of America
The retinoic acid-inducible gene-I (RIG-I)-like receptors are an important family of cytosolic viral RNA sensors. RIG-I recognizes short
5’-triphosphate base-paired viral RNA and is a critical mediator of the innate immune response against viruses such as influenza A, HIV
and hepatitis C. This response requires a carefully orchestrated interaction with tripartite motif 25 (TRIM25). The binding of viral RNA
and the hydrolysis of ATP induce a conformational change in RIG-I, which releases its tandem CARD domains for interaction with the
TRIM25 B30.2 domain. TRIM25 then functions as an E3 ubiquitin ligase to stabilize the formation of a RIG-I tetramer and facilitate RIG-I
interaction with MAVS (mitochondrial anti-viral signalling) at the mitochondrial membrane. The net result is expression of the type I and
III interferon (IFN)s and the establishment of an anti-viral state. We have previously solved the crystal structure of the mouse TRIM25
B30.2 domain and identified key residues that are critical for the interaction with the RIG-I CARDs (Site 1) (1). We have now identified a
second CARD-binding site on the TRIM25 B30.2 domain that is revealed by removal of an N-terminal alpha-helix (mimicking TRIM25
dimerization) (Site 2). We provide biochemical evidence to suggest that both CARDs participate in this interaction and that a
conformational change is required to expose a structurally-similar helix in CARD2. This suggests a model whereby the RIG-I CARDs
interact with opposing sides of the TRIM25 B30.2 domain to form a higher order TRIM25/RIG-I complex that facilitates RIG-I
tetramerisation. The characterization of a dual binding mode for the TRIM25 B30.2 domain is a first for the SPRY/B30.2 family and has
broader implications. For instance, disease-causing mutations in the MEVF gene encoding Pyrin (TRIM20) map to its B30.2 domain in a
region analogous to “Site 2”.
(1) D’Cruz et al., Biochem J. 2013.
308
IL-37 requires IL-18Rα and SIGIRR to carry out its multi-faceted anti-inflammatory program on innate
signal transduction
Claudia A Nold1, 2, Camden Y Lo3, Ina Rudloff1, 2, Suzhao Li4, Michael P Gantier3, Bjoern M Rotter5, Amelie S Lotz6, Soeren W
Gersting6, Tania Azam4, Steven X Cho1, 2, Philip Bufler6, Cecilia Garlanda7, Alberto Mantovani7, Charles A Dinarello4, Marcel F
Nold1, 2
1. Department of Paediatrics, Monash University, Melbourne, VIC, Australia
2. Ritchie Centre, MIMR-PHI Institute of Medical Research, Melbourne, VIC, Australia
3. MIMR-PHI Institute of Medical Research, Clayton, VIC, Australia
4. Department of Medicine, University of Colorado Denver, Aurora, CO, USA
5. GenXPro, Frankfurt, Germany
6. Department of Pediatrics, Ludwig-Maximilians University, Munich, Germany
7. Humanitas Clinical and Research Center, Rozzano, Italy
Aims. Both IL-37 and SIGIRR (TIR8, IL-1R8) are anti-inflammatory orphan IL-1 ligand and receptor family members. Association of
recombinant IL-37 with IL-18 receptor alpha (IL-18Rα) was reported, but no biological activity was observed. Therefore, we investigated
the interactions between IL-37, SIGIRR and IL-18Rα, and their effects on intracellular signaling.
Methods and results. In IL-37-transfected THP-1 macrophages, we observed an 83% reduction in IL-1β, but only 34% when
endogenous SIGIRR was silenced. A similar attenuation of IL-37-induced anti-inflammation was demonstrated in LPS-stimulated human
PBMC, and by IL-18Rα silencing. By immunoprecipitation and immunofluorescence, IL-37 associates with SIGIRR and IL-18Rα in A549
epithelial cells and RAW macrophages, both transfected with IL-37. Using proximity-ligation assays and FRET in PBMC, thus exploring
interactions of the naturally occurring pairs IL-37:SIGIRR, IL-37:IL-18Rα and SIGIRR:IL-18Rα, we demonstrated each pair’s sub-40nm
co-localization. These cell surface-interactions were maximal 30min after LPS. Formation of the three-component ligand-receptor
complex was confirmed by BRET and super-resolution microscopy. We next generated a strain of SIGIRR-KO mice transgenic for IL-37
(IL-37tg-SIGIRR-KO). As expected, IL-37tg mice were protected from endotoxic shock; however, IL-37tg-SIGIRR-KO mice were not:
LPS induced severe hypothermia (trough 25°C) and acidosis (pH7.16) in wild-type, but not in IL-37tg mice (29°C, pH7.32). In IL-37tgSIGIRR-KO mice, these protective effects were considerably weaker. Mechanistical exploration of the effects of the IL-37 receptor
complex on intracellular signaling by proteomic and transcriptomic methods revealed that, via SIGIRR, IL-37 exploits the activities of
anti-inflammatory mediators, e.g. Dok1, and inhibits not only NF-κB and MAPKs, but also mTor and Tak1.
Conclusions. IL-37 engages in a cell surface-complex with IL-18Rα and SIGIRR to limit the severity of inflammation by activating an
intracellular anti-inflammatory program. As it blocks mTor, IL-37 may function as an endogenous rapamycin. SIGIRR utilizes an
unexpected second mechanism of action in addition to the known decoy functions.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 110
309
IL-1 receptor antagonist (IL-1Ra) prevents murine bronchopulmonary dysplasia (BPD) induced by
perinatal inflammation and hyperoxia.
Marcel Nold1, 2, Niamh Mangan3, Ina Rudloff1, 2, Steven Cho1, Nikeh Shariatrian1, Thilini Samarasinghe1, Elizabeth Skuza1, John
Pedersson4, Alex Veldman5, Philip Berger1, Claudia Nold-Petry1, 2
1. The Ritchie Centre, MIMR-PHI Institute of Medical Research , Melbourne, VIC, Australia
2. Department of Pediatrics, Monash University, Melbourne, VIC, Australia
3. Centre for Innate Immunity and Infectious Diseases, Monash Institute of Medical Research, Melbourne, VIC, Australia
4. TissuPath, Mount Waverly, VIC, Australia
5. Monash Institute of Medical Research, Clayton, VIC, Australia
BPD is a common lung disease of premature infants, with devastating short- and long-term consequences. The pathogenesis of BPD is
multi-factorial, but all triggers cause pulmonary inflammation. No therapy exists; thus, we investigated whether the anti-inflammatory IL1Ra prevents murine BPD.
We precipitated BPD by perinatal inflammation (LPS injection to pregnant dams) and rearing pups in hyperoxia (65% or 85% O 2). Pups
were treated daily with IL-1Ra or vehicle.
Vehicle-injected animals in both levels of hyperoxia developed a severe BPD-like lung disease (alveolar number and gas exchange area
decreased by 60%, alveolar size increased 4-fold). IL-1Ra prevented this structural disintegration at 65%, but not 85% O 2. Hyperoxia
depleted pulmonary immune cells by 67%; however, extant macrophages and dendritic cells were hyper-activated, with CD11b and
GR1 highly expressed. IL-1Ra partially rescued the immune cell population in hyperoxia (doubling viable cells), reduced the percentage
that were activated by 63%, and abolished the unexpected persistence of IL-1alpha and IL-1beta on d28 in hyperoxia/vehicle-treated
lungs. On d3, perinatal inflammation and hyperoxia each triggered a distinct pulmonary immune response, with some pro-inflammatory
mediators increasing 20-fold and some amenable to partial or complete reversal with IL-1Ra.
In summary, our analysis reveals a pivotal role for IL-1 in murine BPD and an involvement for MIP-1 and TREM-1. Because it effectively
shields newborn mice from BPD, IL-1Ra emerges as a promising treatment for a currently irremediable disease that may potentially
brighten the prognosis of the tiny preterm patients.
310
Regulation of Th2 cell responses by Grail
Roza Nurieva1, Anupama Sahoo1, Andrei Alekseev1, Lidiya Obertas1
1. MD Anderson Cancer Center, Houston, TX, United States
Aims: T helper (Th)-2 cells are the major players in allergic asthma; however, the mechanisms that control Th2-mediated inflammation
are poorly understood. Our previous studies showed that E3 ubiquitin ligase, Grail, is associated with T cell tolerance. In addition to
tolerant T cells, Grail mRNA is upregulated during normal T cell activation, suggesting that Grail function might not be restricted to T cell
tolerance. In fact, we determined a significant expression of Grail in Th2 cells compared to other T helper subsets, suggesting the role of
Grail in controlling Th2 programming.
Methods: The regulation of Grail expression in Th2 cells was assessed by quantitative Real-Time PCR, Chromatin Immunoprecipitation
(ChIP) assay, luciferase reporter assay and retroviral transduction. Wild-type and Grail deficient T cells were utilized for T helper cell
differentiation assay to evaluate the role of Grail in Th2 responses in vitro. For in vivo studies wild-type and Grail deficient mice were
subjected to Ova immunization and asthma model. Immunoblot analysis and ubiquitination assay were performed to explore the
mechanism whereby Grail controls Stat6 expression in Th2 cells.
Results: In the current study, we found that Grail is selectively induced upon IL-4 stimulation in a time dependent manner and depends
on Th2-specific factors Stat6 and Gata3 that bind to and transactivate the Grail promoter. Grail deficiency in T cells leads to enhanced
Th2 development in vitro and in vivo; Grail deficient mice are more susceptible to allergic asthma. Mechanistically, the enhanced
effector function of Grail-deficient Th2 cells is mediated by increased expression of Stat6 and IL-4 receptor α-chain. Grail interacts with
Stat6 and targets it for ubiquitination and degradation.
Conclusions: Our results suggest an important link between the Th2 specific expression of Grail and its role in control of Th2
developmentand Th2-mediated pathogenesis and immunity through a negative feedback loop.
311
+
CD141 DC and IFN-λ production in the peripheral blood of a young cohort of injecting drug users
(IDUs) with chronic HCV infection
Jeffrey Smith1, Ben Fancke2, 3, Margaret Hellard2, Rosemary Ffrench2, 3, Meredith O'Keeffe2, 3
1. Microbiology and Immunology, University of Melbourne, Melbourne
2. Burnet Institute, Melbourne, VIC, Australia
3. Immunology, Monash University, Melbourne
Polymorphisms within and upstream of Interferon-lambda (IFN-λ) gene 3 (IFN-λ3, IL-28B gene) are strongly associated with the
clearance of Hepatitis C virus (HCV) in chronically infected patients treated with interferon-alpha (IFN-α). Human blood and liver
dendritic cells (DC) expressing CD141 and Clec9A are known to produce IFN-λ in response to dsRNA and in particular, to HCV. In this
study we enumerated the CD141+ DC in the peripheral blood of a young cohort of injecting drug users (IDUs), with chronic HCV
infection, not undergoing treatment, and assayed their ability to produce IFN-λ. DCs from chronic HCV-infected IDUs expressing the
protective IL-28B genotype (rs8099917, TT and rs12979860, CC) were compared to those expressing IL-28B rs8099917, GT or GG and
rs12979860 CT or TT. Regardless of the IFN-λ genotype, IFN-λ production was reduced from the total PBMCs of chronically infected
HCV donors. This reduction was commensurate with a common loss of CD141 + DC from the blood of all chronically infected individuals.
However, the production of IFN-λ was increased at least 2-fold by the pre-treatment of PBMC with IFN-α in healthy subjects and in
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 111
chronically infected patients, regardless of genotype. Thus those cells most efficient at IFN-λ production, CD141+ DC, although
substantially reduced in blood in the cohort we have examined, respond to IFN-α with increased IFN-λ production, regardless of the IL28B genotype.
312
Loss of NF-κB1 Promotes Gastric Cancer through Cytokine Driven Chronic Inflammation
Lorraine A O'Reilly1, Tracy L Putoczki1, Ann Lin1, Matthias Ernst1, Raelene J Grumont2, Lisa Mielke1, Andrew Kueh1, Richard
Ferrero3, Gordon K Smyth1, Yifang Hu1, Steve Gerondakis2, Paul M Waring4, Andreas Strasser1
1. Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
2. Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University Central Clinical School,
Melbourne, Victoria, Australia
3. Monash Institute of Medical Research, Melbourne, VIC, Australia
4. Department of Pathology, University of melbourne, Melbourne, Vic, Australia
Gastric cancer (GC) is the second most common cause of cancer-associated mortality. It has a poor prognosis, with new treatment and
prevention strategies focused on impeding the role of chronic inflammation in disease progression. We present a new mouse model of
invasive GC based on the loss of the NF-κB family member, NF-κB1. We show that loss of NF-κB1 results in the induction of
inflammation (gastritis), gastric atrophy and a series of malignant changes that culminate in invasive gastric adeno-carcinoma.
Importantly, disease manifestation occurs independently of H. pylori infection and even in the complete absence of commensal
microbiota. This is the first GC model that recapitulates all stages of invasive human disease and pertinently, reduced nfκb1 promoter
activity due to polymorphisms/mutations are associated with a significant fraction of human GC in North East Asia
We have generated bone marrow chimeric mice to demonstrate that loss of NF-κB1 in both inflammatory and gastric epithelial cells
(GECs) is required for the development of GC. We also demonstrate that nuclear extracts from gastric epithelial cells of wt mice
predominantly contained NF-κB1 homodimer complexes, which were of course absent from the cells of nfkb1-/-mice. NF-κB1 homodimers are known to associate with Histone deacetylase-1 (HDAC-1) to function as a repressor of NF-κB driven activation of several
inflammatory genes within T cells and macrophages. Pertinently we found elevated levels of several pro-inflammatory cytokines (e.g. IL1β, IL-6, IL-8, IL-17 and TNF-α, all known NF-κB targets) in the sera and GC lesions of nfkb1-/- mice. RNAseq analysis of the lymphoid,
myeloid and epithelial cells isolated from the stomachs of young nfkb1-/-mice identified myeloid cells as the dominant source of these
cytokines. Collectively, our findings demonstrate that NF-κB1 functions as a tumour suppressor in GC and provides anovel model to test
emerging therapeutic strategies for gastric cancer treatment.
313
Sidt2 is required for innate immunity to extracellular dsRNA
Ken Pang1, Tan Nguyen1, Blake Smith1, Alix Weisman2, Seth Masters1, Gabrielle Belz1, Craig Hunter2
1. Walter and Eliza Hall Institute, Parkville, VIC, Australia
2. Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, United States
During viral infection, extracellular double stranded RNA (dsRNA) acts as a potent trigger of innate immunity via the production of type I
interferons (IFNs). The detection of extracellular dsRNA involves different host cell sensors such as Toll-like receptor 3 (TLR3) and the
RIG-I-like receptors (RLRs), RIG-I and MDA-5. TLR3 is located within endo-lysosomes, and is therefore well situated to detect dsRNA
that has been taken up from the environment. Internalized dsRNA also activates the cytoplasmic RLRs, implying the existence of a
mechanism to transport dsRNA across the endosomal membrane. Interestingly, the RLRs are functionally dominant over TLR-3 in the
response to extracellular dsRNA and are critical for immunity to multiple viruses, but how dsRNA escapes from the endosome is
unknown. Here we report that Sidt2, a mammalian ortholog of the C. elegans SID-1 dsRNA transporter, is present within the endosomal
compartment and co-localizes with internalized poly(I:C), a synthetic dsRNA. Moreover, we generated Sidt2-deficient mice and found
that loss of Sidt2 impairs the production of type I IFNs in response to extracellular poly(I:C) both in vitro and in vivo. This impairment in
IFN production was not apparent when testing poly(A:U), a dsRNA ligand that activates only TLR-3 (and not the RLRs), and could be
circumvented by delivering poly(I:C) with transfection reagents designed to facilitate endosomal escape. Together, these observations
suggested a role for Sidt2 in transporting dsRNA from the endosome into the cytoplasm. In support of this, we subsequently found that
loss of Sidt2 does not affect dsRNA internalization, but instead results in the accumulation of poly(I:C) within the endosomal
compartment and a concomitant decrease in signaling events downstream of the RLRs. Finally, we show that Sidt2-deficient mice
produce less type I IFNs and have increased mortality in response to infection with herpes simplex virus. Taken together, our findings
demonstrate a key role for Sidt2 in the endosomal escape of dsRNA and the subsequent induction of innate immunity.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 112
314
Possible role of the NLRP3 inflammasome in the development of human atherosclerosis and
myocardial infarction.
Geena Paramel1, Lasse Folkersen2, Rona Strawbridge2, Pål Aukrust3, karin franzen1, Allan Sirsjö1
1. Orebro University, Department of health and Medical science, Orebro, SE, Sweden
2. Department of Medicine and Center for Molecular Medicine, , Karolinska Institutet, , solna, Stockholm, , Sweden
3. Research Institute of Internal Medicine,Department of Thoracic and Cardiovascular Surgery ,K.G. Jebsen Inflammatory Research
Center, Institue of Clinical Medicine ,Section of Clinical Immunology and Infectious Diseases, University of Oslo, Oslo
Abstract
Objective- The NLRP3 inflammasome is an IL-1β and IL-18 cytokine processing complex that is activated in inflammatory conditions.
The role of the NLRP3 inflammasome in the pathogenesis of atherosclerosis and myocardial infarction (MI) is not fully understood.
Approach and Results- Atherosclerotic plaques were analyzed for transcripts of the NLRP3 inflammasome, and for IL-1β release. The
Swedish FIA cohort consisting of DNA from 555 MI patients and 1016 healthy individuals was used to determine the frequency of 5
SNPs from the downstream regulatory region of NLRP3. Expression of NLRP3, ASC, CASP1, IL1B, and IL18 mRNA was significantly
increased in atherosclerotic plaques compared to normal arteries. The expression of NLRP3 mRNA was significantly higher in plaques
of symptomatic patients when compared to asymptomatic ones. CD68 positive macrophages were observed in the same areas of
atherosclerotic lesions as NLRP3 and ASC expression. Cholesterol crystals and ATP induced IL-1β release from LPS-primed human
atherosclerotic lesion plaques. The minor alleles of the variants rs4266924, rs6672995 and rs10733113 were associated
with NLRP3 mRNA levels in PBMCs and plaques but not with the risk of MI.
Conclusions- Our results indicate a possible role of the NLRP3 inflammasomeand its genetic variantsin the pathogenesis of
atherosclerosis.
315
A type III effector antagonises death receptor signaling during bacterial gut infection
Jaclyn S Pearson1, Cristina Giogha1, Catherine Kennedy1, Giuseppe Infusini2, Andrew Webb2, Gadi Frankel 3, Elizabeth
Hartland1
1. Microbiology & Immunology, University of Melbourne, Parkville, VIC, Australia
2. Proteomics Laboratory, Division of Systems Biology and Personalised Medicine, Walter and Eliza Hall Institute of Medical
Research, Parkville, VIC, Australia
3. Centre for Molecular Microbiology and Infection, Imperial College, London, United Kingdom
Successful infection by enteric bacterial pathogens depends on the ability of the bacteria to colonise the gut, replicate in host tissues
and disseminate to other hosts. Enteropathogenic E. coli (EPEC) is an attaching an effacing (A/E) pathogen that adheres intimately to
the apical surface of host enterocytes and causes acute gastroenteritis in humans. Like other enteric bacterial pathogens such
as Salmonella and Shigella, EPEC utilises a type III secretion system (T3SS) to deliver multiple effector proteins directly into host cells
where they subvert various cellular processes including apoptosis and inflammatory signaling. Given their central role in the
pathogenesis of many bacterial infections, elucidating the biochemical function of T3SS effectors is fundamental to understanding hostpathogen interactions. In this study, we found that the T3SS effector, NleB interrupted host extrinsic apoptotic signaling via the death
receptors, TNFR1 and FAS. NleB expressed ectopically or delivered by the T3SS bound to the death domain (DD) proteins FADD,
RIPK1 and TRADD in a DD dependent manner thereby preventing FasL and TNF-induced caspase-8 activation and cell death.
Furthermore, NleB inhibited the formation of the death inducing signaling complex (DISC) during EPEC infection. This activity was
dependent on the N-GlcNAc transferase activity of NleB1, which specifically modified Arg117 in the death domain of FADD. The
importance of the death receptor apoptotic pathway to host defence was demonstrated using mice deficient in the FAS signalling
pathway, which showed delayed clearance of the EPEC-like mouse pathogen Citrobacter rodentium and reversion to virulence of
an nleB mutant. The activity of NleB suggests that EPEC and other attaching and effacing (A/E) pathogens antagonise extrinsic cell
death pathways to prevent apoptosis of infected cells, thereby interfering with a major antimicrobial host response.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 113
316
The Pyrimidine Synthesis Modulon: A Stabilised Oscillatory Network Governed by Interferon
Signalling.
Andrew Peel1, Tim Shaw2
1. The University of Melbourne, Parkville, VIC, Australia
2. Victorian Infectious Diseases Reference Laboratory, Melbourne, Victoria, Australia
Background
Living cells simultaneously perform multitudes of precise, stringently regulated functions to co-ordinate interactions of the ~30,000 node
gene-protein and protein-protein networks encoded by the genome, a feat achieved with remarkable efficiency (<8 pW per cell), using
imprecise functional units with a large amount of internal redundancy and feedback. At all levels, noisy, overlapping and often
contradictory input signals are integrated and processed to produce stable regulated outputs. Recognition of this striking resemblance to
computing networks has inspired the exploration of a new and expanding area of biological electronics, which aims to use electrical
engineering concepts to emulate complex biological processes.
Defining characteristics of complex biological networks, which have resulted from millions of years of evolutionary experiment, are selforganization, robustness and plasticity. They are stable to fluctuations (noise) in “expected” input, but prone to failure when challenged
by unusually large or prolonged “unexpected” input, to which their response is a self-organized transition to a new meta-stable state, or
failing to achieve that, to extinction. The transition phase is typically scale-free and describable by simple power laws of the form y =
axb. The network becomes refractory to further input and ultimately fails or shuts down if it lacks a required input, or if the amplitude or
frequency of an input signal exceeds a critical threshold.
Modeling and Results
We devised an electronic analogue of the evolutionarily ancient biochemical pathway for de novo pyrimidine synthesis, which requires
six enzymatic reactions, the fourth being coupled to mitochondrial respiration. Rate-limiting steps are directly or indirectly governed by
interferon signaling which regulates flux through the pathway via control of gene expression and gene product stability. The model
predicts that intermittent pulses of interferon stimulation are required to de-repress the pathway and that over-stimulation will cause
mitochondrial “burn-out”, consistent with many published observations.
317
Interferon-β regulates Th17 polarization and dendritic cell migration in EAE
Leesa M Pennell1, 2, Eleanor N Fish1, 2
1. Immunology, University of Toronto, Toronto, ON, Canada
2. Toronto General Research Institute, University Health Network, Toronto, ON, Canada
To investigate the mechansim(s) of action of IFN-β in suppressing immune and inflammatory processes in multiple sclerosis (MS), we
employ a myelin oligodendrocyte glycoprotein (MOG) peptide-induced experimental autoimmune encephalomyelitis (EAE) model of MS
in IFN-β+/+ and IFN-β-/- mice. IFN-β-/- mice exhibit earlier onset and more rapid progression of neurologic impairment compared with IFNβ+/+mice. We provide MRI evidence for a rapid influx of cells into the brains of IFN-β-/- mice with EAE: increased ventricle volume
compared with IFN-β+/+ mice. Th17 cell numbers in the inguinal lymph nodes and in the CNS of IFN-β-/- mice with EAE are higher than
for IFN-β+/+ mice, yet Treg numbers are lower. Anti-CD3/anti-CD28 antibody stimulation of whole splenocytes or CD4+ T cells from IFNβ-/- mice results in production of IL-17A, whereas identical stimulation of cells from IFN-β+/+ mice fails to increase IL-17A production. IFNβ-/- CD4+ T cells express higher levels of IRF-4 following anti-CD3/anti-CD28 antibody activation and increased expression of CCR6, IL23R, IL-6R and CXCR4. Moreover, CD4+ T cells from IFN-β-/- mice exhibit a Th17 primed transcriptome. Increased Th17 cell
polarization during EAE may be driven by dendritic cells (DCs), as DCs derived from IFN-β-/- mice induce greater proliferation of T cells
derived from either IFN-β+/+ or IFN-β-/- mice, compared with DCs derived from IFN-β+/+mice. Additionally, IFN-β-/- DCs secrete cytokines
associated with Th17 rather than Treg polarization. Adoptive transfer of MOG peptide-primed IFN-β-/- DCs into IFN-β+/+ and IFN-β-/- mice
with EAE resulted in their rapid migration into the brains and spinal cords (CNS) of recipient mice, visualized by fluorescence imaging.
FACS analysis of DCs isolated from naïve IFN-β-/- mice revealed increased expression CCR7, CXCR4, and MHCII upon TLR4
activation compared to IFN-β+/+ DCs. Taken together, our data indicate immunoregulatory roles for IFN-β in suppression of Th17 cells
and in limiting the activation and trafficking of DCs during EAE.
318
Targeting chemokines: Pathogens can, why can’t we?
Amanda Proudfoot1
1. Geneva, Switzerland, Geneva, Switzerland
Chemoattractant cytokines, or chemokines, are the largest sub-family of cytokines. About 50 distinct chemokines have been identified in
humans. Their principal role is to stimulate the directional migration of leukocytes, which they achieve through activation of their
receptors, following immobilsation on cell surface glycosaminoglycans (GAGs). Chemokine receptors belong to the G protein coupled
7-transmembrane receptor family, and hence their identification brought great promise to the pharmaceutical industry, since this
receptor class is the target for a large percentage of marketed drugs. Unfortunately, the development of potent and efficacious inhibitors
of chemokine receptors has not lived up to the early expectations. Several approaches to targeting this system will be described here,
which have been instrumental in establishing paradigms in chemokine biology. Whilst drug discovery programs have not yet elucidated
how to make successful drugs targeting the chemokine system, it is now known that certain parasites have evolved anti-chemokine
strategies in order to remain undetected by their hosts. What can we learn from them?
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 114
319
The Interleukin-11 signaling cascade: a promising target for cancer treatment
Matthias Ernst1, Michael Griffin2, Kirsten Edwards3, Tracy Putoczki1
1. Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
2. Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne,
Melbourne, VIC, Australia
3. CSL Ltd., Melbourne, VIC, Australia
Interleukin (IL)-11 is a pleiotropic member of the IL-6 family of cytokines. Despite its discovery over 20 years ago, characterisation of the
function of IL-11 has been over shadowed by its famous sibling IL-6. We have recently solved the first crystal structure of human IL-111,
which signals through specific a-subunit receptors and transmembrane b-subunit GP130 receptors. The formation of the IL-11 signaling
complex results in the recruitment of JAKs and subsequent activation of the pro-tumourigenic transcription factor, STAT3. Constitutive
STAT3 activation is a hallmark of gastrointestinal cancers, and associated with poor prognosis. We have shown that elevated IL-11
expression is linked to high STAT3 activation in human gastrointestinal cancers 2.
In order to compare the requirement of IL-6 and IL-11 signaling for gastrointestinal tumour progression we have coupled il6-/- mice,
which lack both classic and trans-signaling, and il11ra-/- mice with a unique suite of gastric, inflammation-associated and sporadic colon
cancer mouse models. Our results demonstrate that while both cytokines contribute to tumour progression, IL-11 has a dominant role in
the development of gastrointestinal cancers2. We show that the production of IL-11 by cells within the tumour microenvironment
promotes STAT3 activation in non-haematopoietic cells, which triggers the progression of tumours.
We have generated novel IL-11 signaling antagonists and demonstrate in numerous endogenous mouse and human xenograft models
that therapeutic targeting of IL-11 signaling reduces inflammation in the tumour microenvironment and inhibits the progression, invasion
and metastasis of tumour cells. Importantly, inhibition of IL-11 does not result in many of the side-affects commonly associated with
inhibition of other components of the JAK/STAT pathway. Our results highlight an unappreciated hierarchy between IL-6 and IL-11, and
suggest that understanding the cross-talk between cytokines and the tumour microenvironment will prove critical to the design and
success of anti-cytokine reagents as future cancer therapies.
1.
2.
Putoczki et al. (2014) Acta Cryst. D70, doi:10.1107/S1399004714012267
Putoczki et al. (2013) Cancer Cell, 24(2):257
320
Novel insights into interferon regulated expression of the c-MYC gene in human cancer
Samuel J Cutler1, Ibtisam Ghazawi1, James Amalraj1, Glen Boyle2, Grant McArthur3, James Doecke4, Albert Mellick5, Stephen J
Ralph1
1. School of Medical Science, Griffith Health Institute, Griffith University, Gold Coast, QLD, Australia
2. Melanoma Biology, QIMR Berghoffer, Brisbane, QLD, Australia
3. Cancer Research, Peter MacCallum Cancer Research Institute, Melbourne, QLD, Australia
4. Quantitative Biology, CSIRO, Brisbane, QLD, Australia
5. School of Medicine, Faculty of Health, Deakin University, Geelong, VIC, Australia
We have mined datasets from microarray analyses of gene expression from large numbers of cell lines of particular human cancer types
to determine relationships between levels of particular transcription factors and their impact on target gene expression. Using a set of
defined conditions and by excluding confounder data points, this enabled us to then clearly observe significant correlations existing
between sets of transcription factors and their target genes (1). These results were consistently confirmed and validated using other
independent methods for analysis of transcription factor binding, including chromosomal immunoprecipitation, RT-PCR, 5’-RACE, gene
reporter, knockdown of gene expression or gel shift assays. We have applied our microarray analysis of several genes to provide novel
links not previously recognised including some genes involved in, or regulated by the interferon response. For example, IFNg activation
of STAT5 signaling was shown to bind and activate MYC gene expression in human colon cancer. We propose that our approaches
may be more widely applicable, as a general method for refining and determining novel relationships existing between different
transcription factors and the genes that they regulate.
1) REST negatively and ISGF3 positively regulate the human STAT1 gene in melanoma. Amalraj J, Cutler SJ, Ghazawi I, Boyle GM,
Ralph SJ. Mol Cancer Ther. 2013 Jul;12(7):1288-98.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 115
321
Exploiting the type-1 Interferon pathway in the treatment and prediction of breast cancer metastasis
Jai Rautela1, 3, 2, Nikola Baschuk1, Tim Molloy4, Sandra O'Toole4, 6, 5, Paul J Hertzog7, Robin L Anderson3, 2, Belinda S Parker1
1. La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, VIC, Australia
2. Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
3. Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
4. The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW, Australia
5. Sydney Medical School, University of Sydney, Sydney, NSW, Australia
6. Royal Prince Alfred Hospital, Sydney, NSW, Australia
7. Monash Institute of Medical Research, Monash University, Melbourne, VIC, Australia
Metastatic disease accounts for almost all breast cancer related deaths. The immune mechanisms that dictate this spread to key sites
such as the lung and bone are only now emerging. Our recent work emphasizes the role of the host immune system in restricting breast
cancer metastasis, and that the balance between pro- and anti-metastatic immune responses is a critical determinant of metastatic
progression. Previous studies in our laboratory using the syngeneic 4T1.2 breast cancer model have shown that breast tumour cells
suppress their intrinsic type-1 interferon (IFN) pathway, and hence type-1 IFN secretion, in order to evade host immunosurveillance and
metastasise to bone. These findings are supported by a significant correlation between patients with low primary tumour expression of
this pathway and increased risk of bone relapse. Taken together, this highlights the type-1 IFN pathway as an attractive target for novel
anti-metastatic agents.
In order to stimulate a systemic IFN response, we have utilized toll-like receptor (TLR) agonists and are currently evaluating their
potential as anti-metastatic agents. Treatment of tumour bearing animals with the double-stranded RNA mimetic, Poly(I:C), potently
suppressed metastasis to bone and lung and this was associated with enhanced activation of the NK cell compartment. These effects
were superior to that observed by direct administration of IFN alpha. In addition to a closer examination of the cellular and molecular
mechanisms that underpin the efficacy of these anti-metastatic agents, we are investigating the relevance of combining such therapies
with common chemotherapeutics.
Finally, we have uncovered that the expression of a novel set of interferon-regulated genes in a patient’s primary tumour is able to
accurately predict metastasis-free survival. Our data suggest that utilisation of these biomarkers and IFN-based therapies may have a
significant clinical impact in reducing rates of metastatic breast cancer.
We gratefully acknowledge the support of the Cancer Council of Victoria, Sydney Breast Cancer Foundation and the NHMRC of
Australia.
322
Changes in microglial proliferation rate in GFAP-IL10Tg mice following perforant pathway
transection
Mireia Recasens Torné1, Bea Almolda1, Ian Campbell2, Berta Gonzalez1, Bernardo Castellano1
1. University Autonomous of Barcelona, Barcelona, Spain
2. 2School of Molecular Bioscience, University of Sydney, NSW, Sydney
Activation and proliferation of microglia are key events in the response of CNS against inflammatory diseases. Cytokines play an
important role in the control of these processes. Although production of Interleukin-10 (IL-10) by activated astrocytes and microglia has
been demonstrated after different CNS injuries, the specific role played by IL-10 in modulating microglial proliferation remains unclear.
Aims. Hence, the objective of this study was to evaluate the effects of local IL-10 production on microglial proliferation in normal
conditions and associated to axonal anterograde degeneration.
Methods.For this purpose, unilateral perforant pathway transection (PPT) was performed in transgenic mice with astrocyte-targeted
production of IL-10 (GFAP-IL10Tg) and their corresponding wild types (WT) littermates. At 2, 3 and 7 days post-lesion (dpl), animals
were intracardially perfused with 4% of paraformaldehyde and brains processed for immunohistochemical studies. Microglial cell
number was quantified using the myeloid-specific transcription factor PU.1 and microglial proliferation was evaluated using the mitotic
marker Phospho Histone H3 (pH3).
Results. Our results showed a significant increase in the number of microglial cells in GFAP-IL10Tg mice compared with WT, in both
non-lesioned and lesioned animals. The peak of proliferation after PPT lesion was delayed in GFAP-IL10Tg mice showing the maximum
number of pH3+ cells at 3dpl, while in WT the peak was observed at 2dpl. Moreover, the number of proliferating cells was lower in Tg
mice at the different time points analyzed.
Conclusions. In conclusion, this study demonstrated that local production of IL-10 in the CNS modifies the microglial proliferation pattern
associated to PPT. Further studies are necessary to understand the mechanisms involved in microglial proliferation in this paradigm.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 116
323
Role of Bacillus infantis NRRL B-14911 in the mediation of autoimmune myocarditis
Chandirasegaran Massilamany1, Bharathi Krishnan1, Timothy P Smith2, Etsuko Moriyama3, Raul G Barletta1, John D Loy1, Jay
Reddy1
1. School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
2. Genetics, Breeding and Animal Health Unit, U.S. Meat Animal Research Center, Clay Center, NE 68933, USA
3. School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
Environmental microbes can trigger autoimmune responses through multiple pathways, and one mechanism is antigenic mimicry. We
recently reported that a mimicry epitope for cardiac myosin heavy chain (Myhc)-α 334-352, designated BAC 25-40, from Bacillus sp.
NRRL B-14911 can induce myocarditis in A/J mice by generating cross-reactive T cells for cardiac myosin. By 16S rRNA phylogenetic
analysis, we determined the species to be Bacillus infantis NRRL B-14911. Since this microbe was originally isolated from ocean water,
we sought to evaluate the expression of N-carbomoyl L-amino acid amidohydrolase (NCAA), the source protein for BAC 25-40, under
various growth conditions. Unexpectedly, NCAA was found to be expressed in low-salt conditions when bacteria were grown at 37oC,
but not in high salt-containing marine medium, suggesting that the conditions of mammalian systems may favor the expression of NCAA
in this bacterium. Furthermore, we demonstrate that RAW 264.7 cells can phagocytose the bacteria and produce reactive oxygen- and
nitrogen intermediaries to levels comparable to lipopolysaccharide. Likewise, primary macrophages obtained from A/J mice can
produce proinflammatory cytokines, notably, tumor necrosis factor-α, and interleukin-6 in response to bacterial exposure. More
importantly, A/J mice infected with B. infantis show the generation of CD4 T cells that cross-react with both BAC 25-40, and Myhc-α
334-352, as evaluated by major histocompatibility complex class II dextramer staining. Together, the data suggest that B. infantismay
have a potential to induce myocarditis. The establishment of this disease model enables the study of the immune mechanisms of
myocardial injuries of bacterial origin.
324
Serum interleukin 38 is associated with disease severity and organ involvement in systemic lupus
erythematosus.
Ina Rudloff1, 2, Jack Godsell3, Claudia A Nold-Petry1, 2, James Harris3, Alberta Hoi3, Eric F Morand3, Marcel F Nold1, 2
1. Ritchie Centre, MIMR-PHI Institute of Medical Research, Melbourne, VIC, Australia
2. Department of Paediatrics, Monash University, Melbourne, VIC, Australia
3. Centre for Inflammatory Diseases, Monash University, Monash Medical Centre, Melbourne, VIC, Australia
Aims: Interleukin (IL)-38 is a member of the IL-1 cytokine family. Although knowledge on IL-38 is sparse, IL-38 gene polymorphisms
have been associated with inflammatory diseases, and recombinant IL-38 inhibits IL-17 and IL-22. Intriguingly, both IL-17 and IL-22 play
a role in systemic lupus erythematosus (SLE), a severe autoimmune disease. We therefore set out to investigate IL-38 in SLE.
Methods: IL-38 and IL-10 were quantified by ELISA in serum from SLE patients at admission (baseline, 142 patients) and two
subsequent clinic visits (115/142). SLE disease activity index-2000 (SLEDAI-2k) and treatment details were recorded. Serum of 28
healthy donors served as controls. Moreover, we silenced IL-38 in PBMC from healthy volunteers by siRNA (siIL-38) and measured IL-6
by ELISA.
Results: IL-38 (63-5928pg/ml) was detectable in 59 out of 345 patient samples (17.1%) and IL-38 abundance in SLE samples was
significantly higher than in healthy controls (p=0.003). Moreover, patients with active disease (SLEDAI-2k≥4) had 18.4-fold higher serum
IL-38 than patients with non-active disease (p=0.044). Importantly, IL-38 was associated with increased risk of renal (RR=1.6, 95% CI
1.2-2.2, p=0.028) and CNS disease (RR=2.2, 95% CI 1.2-4.3, p=0.035), and when IL-38 was detectable at baseline, patients had a 1.7fold increased risk of developing persistently active disease (RR=1.7, 95% CI 1.3-1.9, p=0.01).Remarkably, siIL-38-treated PBMC from
healthy volunteers produced up to 30-fold more IL-6 than control-transfected cells when stimulated with CpG or imiquimod (n=3).
Similarly, in SLE patients, the anti-inflammatory cytokine IL-10 was 5-fold higher when IL-38 was detectable, suggesting that IL-38 may
be protective in SLE.
Conclusions: This study is the first to demonstrate the anti-inflammatory activity of endogenous IL-38. We moreover reveal IL-38 as the
first mediator that exhibits an association with markers of SLE disease activity and renal and CNS involvement; IL-38 may thus become
the highly sought-after first biomarker for SLE.
325
COLONY-STIMULATING FACTOR-1 AND TUMOR NECROSIS FACTOR- α IN ARTHRTIC PAIN
Reem Saleh1, Andrew Cook1, John Hamilton1
1. Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
Colony-stimulating factor-1 (CSF-1) is a key cytokine that has been linked to the development of arthritis in animal models. Its role in
neuropathic pain has been studied by several laboratories; however, its potential involvement in arthritic pain has not received attention.
Tumor necrosis factor-alpha (TNF-α) is a proinflammatory cytokine which has been implicated in the pathogenesis and progression of
RA, as well as the generation of pain. Thus, the objectives of the current study were as follows: 1) to examine the role of CSF-1 in
arthritic pain using an acute monoarticular methylated bovine serum albumin (mBSA)-induced arthritis model; 2) to compare the TNFmediated pain to the CSF-1-induced pain; and 3) to elucidate the mechanisms by which CSF-1 can mediate arthritic pain. Results from
this study showed that systemic administration of CSF-1 can induce pain in the mBSA model. Preliminary data showed that early CSF1-induced pain was not reversed following indomethacin administration, suggesting that CSF-1-mediated pain is cyclooxygenaseindependent. Systemic administration of TNF-α could also induce pain in the novel mBSA model. Unlike CSF-1, early TNF-mediated
pain was abolished following the administration of indomethacin. This indicated that the TNF-mediated pain is mediated through the
production of eicosanoids. Thus, both cytokines were able to induce pain; however, each cytokine mediated arthritic pain via a different
pathway. Further insight into the mechanisms by which CSF-1 mediates pain could determine its contribution to arthritic pain and may
provide novel therapeutic strategies for joint pain.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 117
326
The RIG-I like receptor pathway: New insights in the subcellular localization of the components
Maite Sanchez-Aparicio2, 1, Juan Ayllon2, 1, Adolfo García-Sastre2, 1
1. Global Health and Emerging Pathogens Institute, New York, USA
2. ICAHN SCHOOL OF MEDICINE AT MOUNT SINAI, NEW YORK, NEW Y, United States
Aims: The RIG-I like receptor (RLR) signaling is an essential pathway for the initiation of host IFN-mediated antiviral responses. The
activation of this pathway is complex and well characterized, but most of the spatio-temporal events, and the subcellular localization
where the essential proteins interact, are still under interrogation.
Methods: The interaction of key partners in the RLR pathway was examined by Bimolecular Fluorescence Complementation (BiFC).
This technique allowed us to isolate, visualize and analyze protein-protein interactions in living cells.
Results: We have defined spatially in the cell different complexes formed between RIG-I, TRIM25 and MAVS, in the presence and
absence of viral IFN antagonistic proteins. Dimers and/or multimers of RIG-I, TRIM25 and MAVS localize into different compartments in
the cytoplasm of the cell. The impact of several viral proteins in the RLR pathway was investigated through BiFC: HCV NS3/4A, IAV
NS1, NiV V and SFTSV NSs proteins. In order to inhibit the IFN response, these viral proteins target specific complexes and interact in
defined areas in the host cell.
Conclusion: For the first time, the interactions among proteins formed along the RLR pathway have been tracked and visualized in
living cells. We found different complexes localized in concrete subcellular compartments not known before. This method has provided
us new information to further understand the mechanisms that regulate the specific distributions of these complexes in the activation of
the RLR pathway.
327
Determining the source of Type III IFNs (IFNλs) in vivo using a novel IFNλ reporter mouse model
Marvin J Sandoval1, Russell K Durbin2, Sergei V Kotenko3, Joan E Durbin2
1. Pathology, NYU School of Medicine, New York, NY, USA
2. Pathology, Rutgers-New Jersey Medical School, Newark, NJ, USA
3. Biochemistry, Rutgers-New Jersey Medical School, Newark, NJ, USA
Similar to Type I IFNs (IFNα/β), Type III IFNs (IFNλ1,2,3,4) exert antiviral and antiproliferative effects through a JAK-STAT mediated
pathway that activates ISGF3 (STAT1/STAT2/IRF9) and consequently induces the expression of interferon-stimulated genes
(ISGs). IFNλs signal through a receptor complex distinct from that of IFNα/β, composed of IL10R2 and IFNλR1, which appears to be
preferentially expressed on epithelial cells and certain immune cell subsets. Accumulating evidence suggests that IFNλs play a major
role in protecting the host from viruses targeting the mucosal surfaces of the respiratory and gastrointestinal tracts as well as the
liver. Like type I IFNs, IFN-λs are produced by various cell types in response to virus infection or TLR ligand stimulation, via
mechanisms that appear to be similar but not identical. The major cell populations contributing to the production of IFNλs in vivo, in
response to specific viral pathogens, remain unclear. To gain a clearer picture of the source(s) of type III IFNs in the course of virus
infection, we have generated a novel IFNλ reporter mouse (IFNλ2+/eGFP), in which one allele of the IFN-λ2 coding sequence has been
replaced with eGFP by homologous recombination, while maintaining the IFN-λ2 promoter regions and UTRs intact. Thus, any cell from
this mouse that produces IFN-λ2 will also express eGFP, allowing us to identify and isolate IFNλ-producing cells present within tissues
as an infection progresses. Preliminary studies using FLT3L and GMSCF cultured, bone marrow-derived dendritic cells from these mice
show that a subpopulation of dendritic cells produce IFN-λ in response to Newcastle disease virus infection, and characterization of
these cells, including their role as type III IFN producers, is now underway. It is anticipated that the availability of the IFNλ2+/eGFP reporter
mouse will help in the identification of IFN-λ producing cells in response to various viral infections in vivo and ultimately aid in
understanding the distinct role IFNλs play in antiviral immunity.
328
Type I IFNs display differential antiviral potency in cells with JAK-STAT signaling deficiencies
William M Schneider1, Hans-Heinrich Hoffmann1, Doron Levin2, Juan L Mendoza3, K. Christopher Garcia3, Gideon Schreiber2,
Charles Rice1
1. The Rockefeller University, New York, NY, United States
2. Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
3. Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California, United States
A multitude of type I IFNs signal through a shared heterodimeric receptor complex. The binding of any one leads to the expression of
hundreds of IFN-stimulated genes that render cells highly resistant to virus infection. Type I IFNs display a marked difference in their
receptor binding affinities, and at varying concentrations, give rise to quantifiable differences in gene induction; however, whether or not
type I IFNs can give rise to gene induction profiles that are qualitatively distinct from one another and whether diverse classes of viruses
are differentially susceptible to their antiviral properties remain unanswered questions. Here, we present data on the characterization of
several type I IFNs and synthetic IFN variants with respect to their effect on gene expression and their ability to restrict diverse viruses.
In physiological settings, biologically important differences in signaling by type I IFNs may arise from variation in the intrinsic abundance
and availability of signaling components. We find that in the presence of high IFN concentrations, STAT1-/- cells are capable of
launching an effective antiviral response; however, the IFNs tested are indistinguishable with respect to antiviral potency. In stark
contrast, in TYK2-/- cells, differences in the antiviral potencies are substantially exaggerated. Despite the large variation in potencies
among the IFNs in different cellular environments, the relative differences are consistent across viruses. These results provide
additional support for a model where a low level of type I IFN signaling is sufficient to elicit a potent pan-antiviral state and suggest that
variations in the level available signaling components may be important for distinguishing type I IFN signaling and fine tuning the IFN
response in biologically relevant cellular environments. In ongoing work, we aim to determine whether distinct gene expression profiles
are induced by type I IFNs in cells with deficiencies in JAK-STAT signaling components.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 118
329
IRF6: a key regulator of oral epithelial cell responses to the human pathogen Porphyromonas
gingivalis
Glen M Scholz1, Jennifer Huynh1, Jiamin Aw1, Mei Qi Kwa1, John A Hamilton1, Eric C Reynolds1
1. University of Melbourne, Parkville, VIC, Australia
Porphyromonas gingivalis is a major pathogen in periodontitis, a prevalent chronic inflammatory disease of the tissues supporting the
teeth and which also affects systemic health. P. gingivalis forms microcolonies in the superficial layers of the polymicrobial biofilm
accreted to the teeth such that it can dysregulate the host immune response to the biofilm resulting in dysbiosis and chronic
inflammation. In addition to functioning as a physical barrier to prevent infection, the epithelial cells abutting the teeth utilize toll-like
receptors (TLRs) to detect and respond to pathogens. The interferon regulatory factor (IRF) transcription factors are key components of
TLR signaling as they impart specificity to downstream responses. In contrast to the other IRFs, IRF6 expression appears to be
restricted to epithelial cells, including those of the oral epithelium. Here, we investigated a role for IRF6 in regulating the responses of
oral epithelial cells to P. gingivalis. The novel IL-1 family cytokine IL-36γ was shown for the first time to be induced in response to P.
gingivalisinfection. IL-36γ expression was also induced by a TLR2 agonist. Gene silencing experiments and gene promoter assays
demonstrated that IRF6 functions downstream of IRAK1 to mediate IL-36γ expression. IL-36γ exerts potent inflammatory effects on
dendritic cells and macrophages, such as induction of the Th17 chemokine CCL20. Significantly, the ability of IL-17 to up-regulate IL36γ expression in oral epithelial cells may further amplify the P. gingivalis-triggered IL-36γ response. IRF6, which we have shown to
also be an important regulator of oral epithelial cell differentiation, is transcriptionally up-regulated in response to P. gingivalis infection.
Collectively, our data therefore indicate that IRF6 functions as an important signaling nexus for pathways regulating both the
inflammatory and barrier functions of oral epithelial cells.
330
Changes in RNA expression pattern in neutrophils of patients with Indian Visceral leishmaniasis
SMRITI SHARMA1, RICHARD DAVIS, SUSANNE NYLEN, MARY WILSON, SHYAM SUNDAR1
1. Department of Medicine, INSTITUTE OF MEDICAL SCIENCES, BANARAS HINDU UNIVERSITY, VARANASI, Uttar Pradesh,
India
Aim- Visceral leishmaniasis (VL), is a disease caused by Leishmania donovani in India. Neutrophils act as first line of defense against
invading microorganisms and their interaction with other immune cells and environmental signals during disease can influence overall
immune response. We observed HLA DR expressing low density "neutrophil like cells" which stain CD66b in PBMC and whole blood
samples in active VL subjects by flowcytometry previously. In continuation to our previous study we also tried to look at changes in
mRNA expression in active VL pre and post treatment (paired) groups and endemic healthy controls (EHC) to understand the
contribution of these cells during active disease.
Methods –
Patient Selection:All patients presented with VL symptoms at KAMRC, Muzaffarpur, India were confirmed to be VL positive by detection
of amastigotes in splenic aspirates and/or by positive rk39 test and EHCs were enrolled from local area.
Gene Expression: Human neutrophils were isolated using Militenyi CD66abce microbead kit to get highly pure neutrophils (> 97%).
cDNA was prepared using High capacity cDNA archive kit. Real time PCR was performed using ABI-Prism 7500 using specific Sybr
Green primers for mRNA of interest.
Results- Differences were observed in expression of cytokine and chemokine mRNAs in the same subjects (paired) in pre and post
treatment(n=8). Some, but not all differences mirrored profiles in EHCs (n=5).
IFN γ,IL10, were significantly up regulated during Active VL while cytokine IL1β and βdefensins and chemokines
CXCL8,CCR4,CCR5,CXCR1,CXCL9 were found to be down regulated. We looked at some important markers like MPO, PD1,
Arginase 1, HLA DR which were found to be upregulated during active VL. These markers were studied to address functional
competency of neutrophils during active VL at mRNA levels.
Conclusions- Neutrophils from subjects with active disease differ from the healthy subjects and from the same subjects after drug
induced cure showing neutrophils during active disease are capable of extensive changes in gene expression and can influence
development of overall immune response.
1.
2.
3.
Richard E davis, University of IOWA and THE VA MEDICAL CENTER, Iowa City, IA, United States.
Mary Wilson, University of IOWA and THE VA MEDICAL CENTER, Iowa City, IA, United States.
Susanne Nylen, Department of Microbiology tumor and cell biology, Karolinska Institutet, Sweden
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 119
331
Interleukin-1 receptor type 2 suppresses collagen-induced arthritis by inhibiting interleukin-1 signal
on macrophages
Kenji Shimizu1, 2, Akiko Nakajima3, Katsuko Sudo3, 4, Yang Liu3, 5, Atsuhiko Mizoroki3, Reiko Horai3, 6, Shigeru Kakuta3, 7, Yoichiro
Iwakura1, 3, 8
1. Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki,
Noda City, Japan
2. Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
3. Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
4. Animal Research Center, Tokyo Medical University, Tokyo, Japan
5. Stem Cell Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
6. Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
7. Department of Biomedical Science, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo, Japan
8. Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama, Japan
Background and aims
Interleukin-1 plays important roles in host defense against infection and inflammatory diseases. IL-1 receptor type 1 is the receptor for
IL-1, and IL-1 receptor type 2 (IL-1R2) is suggested to be a decoy receptor, because it lacks signal transducing TIR domain in the
cytoplasmic part. The roles of IL-1R2 in health and diseases, however, remain largely unknown. Rheumatoid arthritis (RA) is a typical
autoimmune disease affecting approximately 1% of people worldwide independently of races. Because the concentration of IL-1R2
protein is increased in the synovial fluid and plasma in RA patients, the involvement of IL-1R2 in the pathogenesis is suggested. In this
report, we demonstrated the roles of IL-1R2 in the development of arthritis.
Methods and results
We generated EGFP-knock-in Il1r2-/- mice and showed that Il1r2-/- mice were highly susceptible to collagen-induced arthritis, an animal
model for rheumatoid arthritis. Il1r2 was highly expressed in neutrophils but only low in other cells including monocytes and
macrophages. Antibody production and T cell responses against type II collagen were normal in Il1r2-/- mice. In spite of the high
expression in neutrophils, no effects of Il1r2 deficiency were observed in neutrophils. On the other hand, we found that the production of
inflammatory mediators in response to IL-1 was greatly enhanced in Il1r2-/- macrophages.
Conclusion
IL-1R2 is an important regulator of arthritis by acting specifically on macrophages as a decoy receptor for IL-1.
332
Local overproduction of IL10 in astrocyte targeted IL10Tg mice modulates microglial response by
modulating regulatory receptors TREM2 and CD200R following perforant pathway transection.
Kalpana Shrivastava1, Mireia Recasens1, Ian L. Campbell2, Berta Gonzalez1, Bernardo Castellano1
1. Universidad Autonoma de Barcelona, Spain, Barcelona, Spain
2. School of Molecular Biosciences, University of Sydney, NSW, Sydney, Australia
Introduction: Interleukin-10 (IL-10) is a general anti-inflammatory cytokine that has been demonstrated in activated astrocytes and
microglia after CNS injury but its specific role still remains ambiguous. One of the endogenous mechanisms regulating inflammation
following brain injury is the expression of modulator and/or inhibitor membrane receptors. Recent studies demonstrate that TREM2mediated phagocytic function of microglia/macrophages (MM) is required for debris clearance and maintenance of CNS tissue
homeostasis. Moreover, CD200 receptor expressed on microglia is involved in maintaining the microglia in quiescent state.
Aim: We therefore, aimed to study the role of IL10 in modulating microglia using the axonal anterograde degeneration paradigm and to
monitor TREM2 and CD200R expression as an effect of local astrocyte-targeted production of IL-10. We performed a perforant pathway
transection (PPT) on adult GFAP-IL10 transgenic (Tg) mice and corresponding wild types (WT) littermates. Animals were intracardially
perfused at 2,3,7,14 and 21 days post-lesion using 4% paraformaldehyde, brains frozen and sections cut in a cryostat. We performed
single and double immunohistochemistry for microglia expressing TREM2 and CD200R.
Results: Our results show an increase in TREM2 and CD200R expression on microglia in the ipsilateral hemisphere of the Tg and WT
animals especially in the molecular layer of the dennervated dentate gyrus after PPT and the expression was always higher in the Tg
animal. We noticed that their expression is upregulated via local production of IL10 as seen by the activated phagocytosing phenotype
(CD68high and CD16/32high) of microglia in the GFAP-IL10Tg animals. Furthermore, we found a possible feedback inhibition of IL10 as
observed by a constant pSTAT3 expression in Tg mice.
Conclusions: Hence, specific modulation of these receptors and the anti-inflammatory cytokine IL10 with pharmacological tools
represent a promising therapeutic strategy against CNS pathologies. Future studies are concentrated on evaluating the mechanism of
action of IL10 in modulating these regulatory receptors following CNS injury.
333
Epithelial IFN-λ and IFN-α/β constitute a compartmentalized mucosal defense system that restricts
virus infection of the intestinal and respiratory tract
Peter Staeheli1
1. University Medical Center Freiburg, Freiburg, Germany
Not available at time of printing
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 120
334
Strawberry notch homolog 2 is a novel nuclear IL-6 primary response gene in glial cells
Taylor E Syme1, Magdalena Grill1, Stefan Rose-John2, Iain L Campbell1
1. School of Molecular Bioscience & Bosch Institute, University of Sydney, Sydney, NSW, Australia
2. Department of Biochemistry, University of Kiel, Kiel, Germany
Aims: An investigation of potential transcriptional mechanisms underlying interleukin (IL)-6 actions identified a novel putative murine
transcriptional regulator called strawberry notch homolog 2 (Sbno2) as a prominent IL-6 target gene. We hypothesise that Sbno2 is a
key regulator of IL-6 and other gp130 family cytokine actions in the CNS and other tissues. The aim of this study was to better define the
hyper-IL-6-induced regulation of Sbno2 in astrocytes and microglia, as well as to examine its localisation using lentiviral transduction.
Methods: Murine primary cultured astrocytes and the C8-B4 microglial cell line were treated with 25 ng/mL hyper-IL-6 and 10 µg/mL
cycloheximide, and astrocytes were treated with 25 ng/mL hyper-IL-6 and 10 µg/mL actinomycin D. To determine SBNO2 localisation,
lentiviral vectors were produced to deliver an HA epitope-tagged Sbno2 gene and a bicistronic fluorescent marker into the NIH-3T3
murine fibroblast cell line. Fluorescence immunocytochemistry was performed using an antibody against the HA epitope to visualise the
transgenic SBNO2.
Results: Treatment with the protein synthesis inhibitor cycloheximide alone had little to no effect, but superinduced Sbno2 expression
over 8-fold upon hyper-IL-6 treatment in both cell types. Previous results showed that hyper-IL-6-induced Sbno2 expression lasts for at
least 12 h in astrocytes. Inhibition of transcription by actinomycin D revealed Sbno2 mRNA has a half-life of 51 min in these cells. In
NIH-3T3 fibroblasts the transgenic SBNO2 was found to localise exclusively to the nucleus but not the nucleoli.
Conclusions: (1) Sbno2 is a primary response gene to hyper-IL-6 and is able to be regulated by hyper-IL-6 in the absence of additional
protein synthesis, (2) its mRNA levels are tightly regulated, and (3) transgenic SBNO2 localises exclusively to the nucleus consistent
with a role as a transcriptional regulator.
335
Innate cell-like function of T cells in modulation of inflammatory response to infection and tissue
injury
Hong Tang1, 2, Chao Zhang3
1. Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
2. Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
3. Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
Innate cells sense and respond to pathogens, tissue damage and stress by producing pro-inflammatory cytokines and chemokines
(PICC), which are physiological steps to mobilize immune defense, tissue repair and restoration of homeostasis. Abnormal inflammatory
reponses lead to autoimmunity, fibrosis and other autoinflammatory diseases. Thus inflammatory response is tightly controlled both
locally and systemically.
We have previously shown that T cells are both necessary and sufficient to temper Toll like receptor (TLR) mediated inflammatory
response to pathogens, in a TCR-independent manner. TCR-independency thus highlights that T cells may possess evolutionally
conserved characteristics of innate immune cells. Herein I will also discuss another feature of how pro-inflammatory CD8+ T cells
activate macrophages to initiate myocardiac injury casued by hypertension. Macrophages infiltration and activation in myocardium is a
pivotal immunopathological lead to hypertensive cardiac micro-injury, but underlying mechanisms remain elusive. We have found that
CD8+ T cells are essential for the process. CD8 gene targeting (CD8 KO) or CD8+ T cells depletion by antibody significantly reduced
cardiac pro-fibrotic inflammatory responses induced by angiotensin II (Ang II) infusion, whereas CD8 KO mice reconstituted with CD8+
T cells became sensitive to Ang II. More importantly, CD8+ T cells were required for macrophage infiltration in myocardium and
subsequent activation to express PICC. Furthermore, macrophage activation required direct contact with CD8+ T cells, but independent
of T cell receptor (TCR), both in vitro and in vivo. Finally, hypertension may casue myocardium or myofibroblast cells to produce IFNγ,
which is responsible to chemoattract CD8+ T cells to the left ventricles in the onset of cardiac inflammation. Thus, TCR independent
innate nature of CD8+ T cells are both necessary and sufficient to induce hypertensive cardiac inflammation.
In conclusion, innate cells, conventional and non-conventional, seem to be both sensors and effectors that determine the magnitude and
duration of inflammatory response to tissue damage.
336
Characterizing the Molecular Players Involved in Lipopolysaccharide Inflammatory Responses
Elizabeth J Thatcher1, Douglas T Golenbock1
1. U Mass Medical School, Worcester, MA, United States
The human body relies on its ability to sense invading microorganisms in order to mount an effective immune response. A primary
player in the immune systems response to gram-negative bacterial invasion is lipopolysaccharide (LPS). LPS is a critical component of
the outer membrane of gram-negative bacteria, and its endotoxin properties can elicit a large inflammatory response that can induce
sepsis and subsequent death. Presently, we know that the binding of LPS to CD14 initiates signal transduction; however, the molecular
processes immediately following this event remains unclear.
Previously, Chinese hamster ovary (CHO)-K1 cells that stably express CD14 were mutagenized and exposed to LPS. Two
complementation groups were isolated from this screen, both of which fail to response to LPS stimulation but still respond to TNF
stimulation. Complementation Group A was further characterized and found to contain mutations in MD-2. Soluble MD-2 has since been
shown as a critical regulator of cellular responses to LPS through TLR4 signaling. Here, I have utilized whole transcriptome sequencing
to characterize all mutations in Complementation Group B. I am currently using wild-type expression constructs isolated from the other
complementation group to verify the gene required to induce a LPS non-responsive phenotype in Complementation Group B. After in
vitro confirmation, these mutations will be incorporated into engineered mouse models in order to characterize the in vivo response of
this mutation.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 121
337
Characterisation of human metapneumovirus infection of primary human bronchial cells
Belinda J Thomas1, 2, Penny A Rudd3, Suresh Mahalingam3, Philip G Bardin1, 2
1. Monash Lung and Sleep, Monash Medical Centre, Melbourne, Vic, Australia
2. MIMR-PHI Institute, Clayton, VIC, Australia
3. Institute for Glycomics, Griffith University, Brisbane, QLD, Australia
Aims: Human metapneumovirus (HMPV) is a recently discovered respiratory pathogen that presents with similar clinical indications to
respiratory syncytial virus (RSV), ranging from mild respiratory symptoms to severe cough, wheezing, bronchiolitis and pneumonia.
There is mounting evidence suggesting an association between HMPV infection and asthma exacerbation in both adults and children.
Our study examines HMPV infection of primary human bronchial airway cells, comparing responses from normal and asthmatic patients.
Methods: We infected primary human bronchial epithelial cells, fibroblasts and macrophages with HMPV and examined viral replication,
production of inflammatory mediators and anti-viral responses.
Results: HMPV can infect human bronchial epithelial cells, fibroblasts and macrophages; however, we have observed several
differences between the cell types. Viral replication peaks at 48 hours post infection in epithelial cells, whereas replication is still
increasing at 72 hours post infection in fibroblasts. Epithelial cells express MIG, whereas fibroblasts and macrophages release MCP-1.
All cell types have a strong anti-viral response, expressing IP-10 and RANTES and the interferon-stimulated gene, ISG56.
Conclusions: Our data demonstrate that HMPV infects and replicates in primary human bronchial epithelial cells, fibroblasts and
macrophages, and induce a strong inflammatory and anti-viral response. Future studies are looking at these indices in cells obtained
from asthmatic patients to identify a mechanism to explain the link between HMPV infection and asthma etiology and exacerbation.
338
A flow cytometric assay for ASC speck formation in inflammasome responses
Sara J Thygesen1, David Sester1, Vita Sagulenko1, Jasmyn Cridland1, Geoff Osborne2, Kate Schroder3, Kate Stacey1
1. School of Chemistry and Molecular Biosciences , The University of Queensland, Brisbane, Queensland, Australia
2. Queensland Brain Institute, The University of Queensland , Brisbane, Queensland , Australia
3. Institute for Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
Inflammasomes are protein complexes that form in response to infection, cell damage and environmental stress. Inflammasome
activation leads to the processing of proinflammatory cytokines and pyroptotic cell death through the recruitment and activation of
caspase 1, and apoptosis via caspase 8 activation. A common component of many inflammasomes is the adaptor molecule “apoptosis
associated speck-like protein containing a caspase activation recruitment domain” (ASC). ASC is normally spread throughout the cell,
however, upon inflammasome activation the vast majority of ASC is recruited into a single dense speck in the cytosol. We considered
that the dramatic relocalisation of ASC should be detectable by flow cytometry. An assay has been developed that differentiates cells
with ASC specks from cells with diffuse ASC by comparing time of flight parameters (height, width and integral/area) of the fluorescence
pulse generated as a cell passes through the laser beam. This analysis has been applied to native ASC within immunostained
macrophages and monocytes, as well as to ASC-GFP overexpressed in HEK293 cells. Applications for this method of speck detection
include examination of early events in inflammasome formation, and detection of inflammasome responses within mixed cell populations
using lineage markers. In addition this will allow dissection of structural requirements for ASC speck formation using cells reconstituted
for inflammasome formation.
339
Mast cell differentiation is operated by a T cell response
Shota Toyoshima1, Yuuki Obata1, Ei Wakamatsu1, Ryo Abe1
1. Tokyo University of Science, Noda City, CHIBA, Japan
After exposure to an allergen, mast cells (MCs) accumulate at the allergic site and then act as effector cells of inflammation. Despite
many investigations into of the role of MCs in allergic inflammation, the mechanism underlying the expansion of these cells under
allergic inflammatory conditions remains unclear.
When splenocytes from mice immunized with OVA or KLH were repeatedly stimulated in vitro with antigens, expansion of MCs was
greater than that of antigen-specific T cells for up to 3 weeks. This observation raised the possibility that T cell responses elicited by the
antigens were associated with MC development. To verify this, purified splenic T cells from immunized mice were cocultured with MCs
or various progenitor populations and repeatedly stimulated with antigens. The results obtained from these coculture experiments
suggested that T cell responses could have an effect on myeloid progenitors and act to drive the MC differentiation. Additionally, IL-3
from T cells was crucial for MC differentiation. To validate whether MC differentiation that we observed in in vitro culture experiments
might also occur in allergic disease, we used a murine food allergy model that was dependent on T cells and displayed an accumulation
of MCs the colon, and found that this murine model exhibited the accumulation of myeloid progenitors and the up-regulation of IL-3.
Collectively, our study suggests that T cell responses against allergens may drive MC differentiation to generate effector cells of
allergic inflammation. Blocking this differentiation pathway may be an attractive therapeutic strategy for treating allergic inflammation.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 122
340
Apoptotic Cell Recognition Receptors, TYRO3, AXL and MER (TAM), Demonstrate Distinct Patterns
and Complex Regulation of Ligand-Induced Activation and Signaling
Wen-I Tsou1, Khanh-Quynh N Nguyen1, Daniel A Calarese2, Scott J Garforth2, Anita Antes1, Sergey V Smirnov1, Steve C Almo2,
Raymond B Birge1, Sergei V Kotenko1
1. Rutgers, New Jersey Medical School, Newark, NJ, United States
2. Albert Einstein College of Medicine of Yeshiva University, New York, NY, USA
Efficient recognition and removal of apoptotic cells by phagocytes lead to the active suppression of inflammatory responses and the
induction of tolerance. Apoptotic cell (AC) recognition receptors, TYRO3, AXL and MER, share similar structural organization of their
extracellular and intracellular domains, and form a subfamily of receptor tyrosine kinases (RTKs), abbreviated as TAMs. Activation of
TAMs is triggered by secreted glycoproteins, growth arrest-specific gene 6 (GAS6) or protein S (PROS1), that bind to TAMs through
their C-terminal Laminin G (LG) domains. In turn, GAS6 and PROS1 are γ-carboxylated on glutamic acid residues in their N-terminal
Gla domains and interact in a Ca2+-dependent manner with externalized phosphatidylserine (PS) on the surface of ACs and enveloped
viruses. Therefore, GAS6 and PROS1 act as bridging factors: their γ-carboxylated Gla domains bind to PS and opsonize ACs, whereas
their LG domains interact with TAMs, enabling TAMs to bind indirectly to ACs and act as immunoregulatory receptors that dampen
inflammation. Although TAMs share significant similarity, very little is known about the specificity of interaction between TAMs and their
ligands in the context of apoptotic cells, and about downstream signaling cascades. We generated and utilized a set of reporter cell lines
expressing chimeric TAM receptors to demonstrate that each TAM has a unique pattern of interaction with GAS6 and PROS1. These
interactions are also differentially affected by the presence of ACs, PS liposomes, and enveloped viruses. We also demonstrated that γcarboxylation of ligands is essential for the full activation of TAMs. In addition, we found that TAM-mediated signaling up-regulates
expression levels of programmed death-ligand 1 (PD-L1) and PD-L2 that play an important role in immunosuppression. Overall, these
studies reveal that, despite their similarity, TYRO3, AXL and MER perform distinct but overlapping functions in the recognition and
removal of ACs, and in the regulation of inflammatory and immune responses.
341
Poor outcome in rheumatoid arthritis patients switching from etanercept to tocilizumab therapy
Kazuko Uno1, Kazuyiki Yoshizaki2, Michihiro Iwahashi3, Miki Tanigawa2, Katsumi Yagi1
1. Louis Pasteur Center for Medical Research, Kyoto City, KYOTO, Japan
2. Department of Immuno-medical Science, Division of Applied Chemistry, Graduate School of Engineering, , Osaka University, Suita,
Osaka, Japan
3. Higashi Hiroshima Memorial Hospital, Higashi Hiroshima, Hiroshima, Japan
At present, there is no standard by which to pre-qualify rheumatoid arthritis (RA) patients for tocirizumab therapy among those who have
failed previous anti-cytokine therapy (non-naïve). In this study, forty-one biologic non-naïve patients who had failed one to three prior
treatments with methotrexate combined with anti-TNF-a therapy were enrolled in this study. Prior to receiving tocilizumab treatment,
serum cytokine, chemokine and soluble receptor levels were measured using Bio-Plex human cytokine/chemokine 27-Plex panel and
Millipore soluble receptors panel (sIL-6R, sgp130, sTNFR-I and sTNFR-II). Patients were treated with 8mg/kg of tocilizumab once every
4 weeks. After 16 weeks of therapy (4 administrations), clinical efficacy was judged based on patients’ disease activity score (DAS28CRP), and whether patients experienced complete remission or non-remission.
All non-naive RA patients except one showed improvements in their DAS28-CRP score at week 16 of therapy and 9 out of 40 (22.5%)
non-naïve patients experienced clinical efficacies that were judged as complete remission. The remaining 31 patients experienced nonremission (symptom levels: low=6, median=21, and high=4). In the same study, complete remission (DAS-28-CRP≧2.3) was achieved
by 56 % of naïve patients. Although 20 out of 21 patients who switched form etanacept to tocirizumab had improved DAS28 score only
two of these patients achieved remission. On the other hand, 5 of 11 patients switching from infliximab reached remission. Single
logistic regression analysis revealed that sgp130 and sIL-6R level played a role in these outcomes.
Additionally in comparing naïve and non-naïve RA patients’ pre-treatment cytokine/chemokine levels, we observed that they were
relatively similar except for sTNFR-II, which was higher than in non-naïve patients.
These results suggest that prior RA treatment may influence the outcome of further treatments using different biological drugs. Using
prediction biomarkers may help clinicians to select more treatments with more favorable outcomes especially for patients who are
switching from one treatment protocol to another.
342
Procaspase-8 recruitment and activation at the AIM2 inflammasome
Parimala Vajjhala1, Siew W Pang1, Vitaliya Sagulenko1, David Sester1, Simon Cridland1, Justine Hill1, Katryn Stacey1
1. School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
Inflammasomes are macromolecular complexes that mediate inflammatory and cell death responses to pathogens and cellular stress
signals. Inflammasome formation leads to activation of both caspase-1 and caspase-8. We have studied the molecular interactions
important for assembly of the AIM2 inflammasome and for procaspase-8 recruitment and activation. AIM2 is a member of the PYHIN
(pyrin domain (PYD) and HIN domain-containing) family and is activated in response to cytosolic DNA. Upon activation, AIM2
oligomerises and recruits the adaptor protein ASC. ASC is composed of an N-terminal PYD, with which it mediates a homotypic
interaction with AIM2 PYD and a C-terminal CARD domain, with which it recruits procaspase-1. We show that procaspase-8 interacts
with both ASC and AIM2 via interactions between the tandem DEDs of procaspase-8 and the PYDs of ASC and AIM2. Mutations on
ASC PYD that disrupt its self-association also disrupt interaction with AIM2 and procaspase-8 suggesting that that either ASC PYD selfassociation is important for the different interactions or that the interaction mode between the three domains is similar. However,
swapping of the PYDs between ASC and AIM2 disrupted inflammasome function arguing against identical surfaces being used for all
three interactions. Furthermore, we show that procaspase-8 is efficiently recruited to specks formed by ASC alone but is only activated
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 123
in specks containing both ASC and AIM2. This indicates a more stringent requirement for procaspase-8 activation than for its
recruitment. The inflammasome does not just cluster caspases, but also promotes a specific orientation of procaspase-8 that permits
activation.
343
DYSTONIA-CAUSING FRAME-SHIFT MUTATION IN PACT ACTIVATES INTERFERON-INDUCED
PROTEIN KINASE PKR BY AGGREGATE FORMATION
Lauren S Vaughn1, Rekha C Patel1
1. University of South Carolina, Columbia, SC, United States
PKR or Protein Kinase R is an interferon-induced, double stranded RNA (dsRNA)-activated eIF2α kinase that is a key regulator of
cellular stress response pathways. PACT (also known as PRKRA, and DYT16) activates PKR in response to oxidative stress,
endoplasmic reticulum (ER) stress, and serum starvation. Prolonged PKR activation in response to cellular stress leads to cell death by
apoptosis. An inherited form of early-onset dystonia has been identified to be associated with multiple mutations in the coding region of
PACT. Dystonia is a neurological disorder defined by an occurrence of sustained involuntary muscle contractions, often leading to
disabling and abnormal postures. Several recessive missense mutations and one dominant frame-shift mutation in PACT have been
identified to result in dystonia. A German patient was recently identified as having a heterozygous mutation in exon 3 of PACT (AT
deletion at c.266-267) creating a premature stop codon, resulting in a truncation at AA position 109. The main hypothesis of this
research is that the frame-shift mutation in PACT leads to production of a truncated protein that is misfolded and forms aggregates
resulting in cellular stress and the disease phenotype. We compared the ability of wild type PACT and frame-shift (FS) PACT proteins to
interact with and activate PKR using in vitro biochemical assays. In addition, we have investigated changes in sub-cellular localization,
protein aggregation, and cell death resulting from FS PACT mutant. Our results indicate that although FS PACT lacks the PKR
activation domain, it activates PKR robustly in vitro because it forms aggregates. In addition, the truncated FS mutant protein also
exhibits altered sub-cellular localization and forms aggregates in mammalian cells. These results raise a possibility that the PKR
activation observed in several neurodegenerative diseases that are caused by protein aggregation may be an important contributing
factor for the disease pathology.
344
Guanylate-Binding Proteins (GBPs) and cytoskeleton regulation
Deborah Vestal1, Angela Messmer-Blust1, Suzan Wadi1, Sujata Balasubramanian1, Jonathan Jeyaratnam1
1. University of Toledo, Toledo, OH, United States
Guanylate-Binding Proteins (GBPs) are a family of pro-inflammatory cytokine-induced genes. These proteins have been best studied
for their anti-microbial activities against pathogens sensitive to IFN-gamma. However, GBPs have a number of significant functions not
directly related to their antimicrobial activities. One of these is regulation of the cytoskeleton. Integrins, TNF-alpha, and PDGF all
promote rearrangement of the actin cytoskeleton, in part through the activation of the master cytoskeleton regulator Rac. mGBP-2
inhibits these rearrangements by inhibiting their ability to activate Rac. The inhibition of Rac is accompanied by the binding of mGBP-2
to the catalytic subunit of PI3-K and inhibiting its function. mGBP-2 also protects cells from killing by the cytotoxic chemotherapeutic
drug, paclitaxel. Paclitaxel is proposed to kill cells by inhibiting the dynamic instability of microtubules. Cells can be protected from
paclitaxel by restoration of this dynamic instability. Forced expression of the human ortholog of mGBP-2, hGBP-1, also protects tumor
cells from killing by paclitaxel. hGBP-1 mRNA is expressed at greater than 2-fold higher levels than in benign ovaries in only 11% of
new ovarian tumors. This number was at least 40% for tumors that recurred after a therapeutic regime containing a taxane. While the
mechanism(s) by which hGBP-1 protects cells from paclitaxel is still under investigation, one possibility is that this occurs through
hGBP-1-mediated alterations in cytoskeletal dynamics.
345
ASTROCYTE-TARGETED PRODUCTION OF IL-10 INDUCES CHANGES IN MICROGLIAL REACTIVITY
AND REDUCES MOTOR NEURON DEATH AFTER FACIAL NERVE AXOTOMY
Nàdia Villacampa1, Beatriz Almolda1, Antonietta Vilella2, Iain Campbell3, Berta Gonzalez1, Bernardo Castellano1
1. University Autonomous of Barcelona, Bellaterra, Spain
2. Department of Biomedical, Metabolic and Neural Sciences, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
3. School of Molecular Bioscience, University of Sydney, Sydney, NSW, Australia
Interleukin-10 (IL-10) is a cytokine that plays a crucial role in regulating the inflammatory response and immune reactions. In the central
nervous system (CNS), IL-10 is mainly produced by astrocytes and microglia and its upregulation has been demonstrated after various
insults, such as experimental autoimmune encephalomyelitis, middle cerebral artery occlusion, excitotoxicity and traumatic brain injury.
To better understand the effects of IL-10 in the normal and injured CNS, we generated transgenic mice expressing the murineIL10 gene
under the transcriptional control of the GFAP promoter (GFAP-IL10Tg). Previous studies by our group demonstrated specific changes in
microglial phenotype under basal conditions in GFAP-IL10Tg mice. The objective of the present study was to investigate the effects of
local astrocyte-targeted IL-10 production on microglial activation, neuronal degeneration and leukocyte recruitment after experimental
axotomy. Our results showed that after facial nerve axotomy, GFAP-IL10Tg mice showed remarkable changes in the expression of
different molecules associated with the microglial activation pattern, such as CD16/32, CD18 and MHC-II, as well as in the number of
microglial clusters and in microglial cell density. Furthermore, a higher CD3+ lymphocyte infiltration was observed on the axotomized FN
of GFAP-IL10Tg mice. These changes in microglial activation and lymphocyte recruitment correlated with a reduced motor neuron death
at 21 dpi. Altogether, our findings suggest that astrocyte-targeted production of IL-10 impacted the microglial response and lymphocyte
recruitment and culminated in a beneficial effect on neuronal survival.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 124
346
Cytoplasmic DNA induces rapid cell death in Drosophila cells
Nazarii Vitak1, Karyn Johnson2, David P Sester1, Katryn J Stacey1
1. School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
2. School of Biological Sciences, University of Queensland, Brisbane, QLD, Australia
Eukaryotic cells sequester their DNA in the nucleus and organelles, leaving cytosol DNA-free. The presence of DNA in the cytosol
indicates a danger, such as infection, activity of retrotransposons or DNA damage. We hypothesized that defence against cytosolic DNA
is a feature of all eukaryotic cells, and fundamental to maintenance of genome integrity as well as recognition of infection. In mammals
cytosolic DNA is recognized by AIM2, a member of the HIN-200 family, leading to formation of a multimolecular inflammasome complex,
that in turn induces a rapid lytic death in macrophages, termed pyroptosis. There is no data about such a system for invertebrates, and
AIM2 itself is a mammalian-restricted protein. Moreover there is no indication of active necrotic cell death in insects. To investigate
whether an analogous system operates in Drosophila, cells were transfected with different types of DNA via electroporation and viability
was assessed either by MTT assay or by exclusion of membrane-impermeable DNA stains. We have found that both primary
hemocytes, and the Drosophila S2 macrophage-like cell line are sensitive to cytosolic DNA. Results showed that DNA from various
sources, but not synthetic double stranded RNA caused rapid cell death. Unlike mammalian DNA-dependent cell death, denatured DNA
was also toxic. Experiments with synthetic DNA showed that the response was sequence-specific. The mode of cell death was
investigated using flow cytometry. Rapid loss of membrane integrity, lack of a population of cells with sub-G0/G1 levels of DNA, and
insensitivity to the pancaspase inhibitor z-VAD-fmk showed that cell death was not apoptotic. We conclude that sensing of cytosolic
DNA in fruitfly induced a novel lytic pathway of cell death. Elucidation of the molecular basis for this DNA recognition will allow
determination of whether it plays a role in defence against infection or in guarding genome integrity.
347
Regulation of inflammatory cytokine responses by a new TLR4 regulator complex of Rab8a/PI3Kγ
Adam A Wall1, Lin Luo1, Jeremy C Yeo1, Nicholas D Condon1, Jennifer Stow1
1. University of Queensland, St Lucia, QLD, Australia
Introduction: Toll-like receptor 4 (TLR4) is activated by bacterial lipopolysaccharide (LPS) to mount innate immune responses that
involve the initial release of pro-inflammatory cytokines, followed by the regulator, or anti-inflammatory, cytokines. Failure to constrain
inflammatory responses results in acute and chronic inflammatory disease. Multiple molecular mechanisms are emerging to control the
cytokine profile that is released after receptor activation.
Results: Here we find TLR4 complexes localised in LPS-induced dorsal ruffles on the surface of macrophages. The small GTPase
Rab8a is similarly enriched in these membrane domains. By using mass spectroscopy and biochemical mutational analysis, we have
established a direct interaction between Rab8a and the phosphatidylinositol 3-kinase γ (PI3Kγ). Knockdown of Rab8a or PI3Kγ,
pharmacological inhibition, or genetic ablation of PI3Kγ all serve to reduce Akt signalling in response to LPS/TLR4. The cytokine profiles
in cells depleted of Rab8a and mice lacking PI3Kγ were examined and we show that in both cases there is a dramatic increase in the
production of the pro-inflammatory cytokines IL-6 and IL-12p40 with a concomitant decrease in the production of the anti-inflammatory
cytokine IL-10. This was mirrored by inhibition of mTORC1 by rapamycin, illustrating that Rab8a/PI3Kγ function as a novel amplifier of
the Akt/mTOR pathway downstream of TLR4.
Conclusions: Our studies introduce Rab8a and PI3Kγ as a novel complex and provide the first biochemical evidence for a Rab
interacting with the immune-specific class IB PI3K. This positions PI3Kγ in a TLR4 cross-talk pathway that may serve as a
pharmacologically relevant target during the innate immune response.
348
Regulation of embryonic hematopoiesis by a cytokine inducible SH2-domain (CISH) homologue in
zebrafish
Rowena S Lewis1, Suzita M Noor1, Fiona W Fraser1, Robert Sertori1, Clifford Liongue1, Alister C Ward1
1. School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
Cytokine-inducible SH2-containing protein (CISH), a member of the suppressor of cytokine signaling (SOCS) family of negative
feedback regulators, is induced by cytokines that activate signal transducer and activator of transcription 5 (STAT5) and can inhibit
STAT5 signaling in vitro. However, demonstration of a definitive in vivo role for CISH during development has remained elusive. This
study employed expression analysis and morpholino-mediated knockdown in zebrafish in concert with bioinformatics and biochemical
approaches to investigate CISH function. Two zebrafish CISH paralogues were identified, cish.a and cish.b, with high overall
conservation (43-46% identity) with their mammalian counterparts. The cish.a gene was maternally derived, with transcripts present
throughout embryogenesis, and increasing at 4-5 days post fertilization, while cish.b expression commenced at 8 hours post fertilization.
Expression of cish.a was regulated by the JAK2-STAT5 pathway via conserved tetrameric STAT5 binding sites (TTCN3GAA) in its
promoter. Injection of morpholinos targeting cish.a – but not cish.b or control morpholinos – resulted in enhanced embryonic
erythropoiesis, myelopoiesis and lymphopoiesis, including a 2-3 fold increase in erythrocytic markers. This occurred concomitantly with
increased activation of STAT5. This study indicates that CISH functions as a conserved in vivo target and regulator of STAT5 in the
control of embryonic hematopoiesis.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 125
349
Critical role of cytokine and growth factor receptors in AML identified using an RNAi functional
screen
Kevin M Watanabe-Smith1, 2, 3, Ryan J MacKenzie1, 3, Christopher A Eide1, 3, 4, Alyssa B Carey1, 3, Monika A Davare5, 6, Cristina E
Tognon3, 4, Rita M Braziel7, Jeffrey W Tyner3, 8, Brian J Druker1, 3, 4, Anupriya Agarwal1, 3
1. Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR, United States
2. Cancer Biology Program, Oregon Health and Science University, Portland, OR, United States
3. Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
4. Howard Hughes Medical Institute, Portland, OR, United States
5. Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
6. Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
7. Department of Pathology, Oregon Health and Science University, Portland, OR, United States
8. Department of Cell & Developmental Biology, Oregon Health and Science University, Portland, OR, United States
Methods: Acute myeloid leukemia (AML) is one of the most common hematologic malignancies affecting both children and adults.
Because cytokine and growth factor receptors contribute to cancer pathogenesis by regulating growth, differentiation, and survival1 2 3 ,
we designed a functional RNAi screen targeting 188 cytokine and growth factor receptors that we found highly expressed in primary
leukemia specimens. Using this screen we identified interleukin-2 gamma receptor (IL2Rγ) and Colony Stimulating Factor 2 Receptor
Beta (CSF2RB)as critical growth determinants for JAK3 mutation-positive AML cells.
Results: We observed that knockdown of IL2Rγ reduced the growth of JAK3 mutation-positive AML cells and abrogates
phosphorylation of JAK3 and downstream signaling molecules, JAK1, STAT5, MAPK and pS6 ribosomal protein. Overexpression of
IL2Rγ in murine cells increased the transforming potential of activating JAK3 mutations, whereas absence of IL2Rγ completely
abrogated the clonogenic potential of JAK3A572V mutation-positive cells. In addition, mutation at the IL2Rγ interaction site in the FERM
domain of JAK3 (Y100C) completely abrogated JAK3-mediated leukemic transformation. Mechanistically, IL2Rγ and JAK3 contribute to
constitutive JAK/STAT signaling through their reciprocal regulation.
Additionally, CSF2RB was identified as a secondary hit in CMK cells and siRNA-mediated knockdown reduced cellular viability and
phosphorylation of JAK2, STAT5, ERK1/2 and S6 ribosomal protein. CSF2RB is highly expressed in 30% of AMLs4 and preliminary
data shows some primary AMLs may depend on CSF2RB expression, indicating a potential therapeutic target.
Conclusions: We have demonstrated: 1) A novel role for IL2Rγ in potentiating oncogenesis in the setting of JAK3-mutation-positive
leukemia. 2) CSF2RB is a potential functional target in AML and future studies investigating this possibility may yield novel therapeutic
strategies. 3) RNAi-based functional assays can be used to facilitate the identification of non-kinase cytokine and growth factor receptor
targets for inhibiting leukemic cell growth.
1.
2.
3.
4.
Graf M, Hecht K, Reif S, Pelka-Fleischer R, Pfister K, Schmetzer H. Expression and prognostic value of hemopoietic cytokine
receptors in acute myeloid leukemia (AML): implications for future therapeutical strategies. Eur J Haematol. 2004 Feb;72(2):89106. PubMed PMID: 14962246. eng.
Van Etten RA. Aberrant cytokine signaling in leukemia. Oncogene. 2007 Oct 15;26(47):6738-49. PubMed PMID: 17934482.
Vainchenker W, Constantinescu SN. JAK/STAT signaling in hematological malignancies. Oncogene. 2013 May 23;32(21):2601-13.
PubMed PMID: 22869151.
Riccioni R, Diverio D, Riti V, Buffolino S, Mariani G, Boe A, Cedrone M, Ottone T, Foa R, Testa U. Interleukin (IL)-3/granulocyte
macrophage-colony stimulating factor/IL-5 receptor alpha and beta chains are preferentially expressed in acute myeloid leukemias
with mutated FMS-related tyrosine kinase 3 receptor. Br J Haematol. 2009 Feb;144(3):376-87. PubMed PMID: 19036083
350
Syk signaling in dendritic cells orchestrates innate resistance to systemic fungal infection
Paul G Whitney2, 1, Eva Bär3, Fabiola Osorio1, Neil c Rogers, Barbara U Schraml1, Safia Deddouche1, Salomé LeibundGutLandmann3, Caetano Reis e Sousa1
1. Immunobiology Laboratory, Cancer Research UK - London Research Institute, London, United Kingdom
2. Doherty Institute, Melbourne, VIC, Australia
3. Institute of Microbiology, ETH Zurich, Zürich, Switzerland
Host protection from fungal infection is thought to ensue in part from the activity of Syk-coupled C-type lectin receptors and MyD88coupled toll-like receptors in myeloid cells, including neutrophils, macrophages and dendritic cells (DCs). Given the multitude of cell
types and receptors involved, elimination of a single pathway for fungal recognition in a cell type such as DCs, primarily known for their
ability to prime T cell responses, would be expected to have little effect on innate resistance to fungal infection. Here we report that this
is surprisingly not the case and that selective loss of Syk but not MyD88 in DCs abrogates innate resistance to acute systemic Candida
albicans infection in mice. We show that Syk expression by DCs is necessary for IL-23p19 production in response to C. albicans, which
is essential to transiently induce GM-CSF secretion by NK cells that are recruited to the site of fungal replication. NK cell-derived-GMCSF in turn sustains the anti-microbial activity of neutrophils, the main fungicidal effectors. Thus, the activity of a single kinase in a
single myeloid cell type orchestrates a complex series of molecular and cellular events that underlies innate resistance to fungal sepsis.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 126
351
The role of PTPN2 in early thymocyte development and JAK/STAT signaling
Florian Wiede1, Jarrod A. Dudakov2, Catherine J. van Vliet3, Dale I. Godfrey3, Richard Boyd4, Tony Tiganis1
1. Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
2. Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, United States of America
3. Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC, Australia
4. Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
The role of common -chain receptor cytokines in the promotion of thymocyte development and peripheral T cell survival is well
characterized. However, the mechanisms by which cytokine signaling is attenuated during T cell development, particularly at the doublenegative (DN) stages, remain unclear. We have shown previously that the protein tyrosine phosphatase PTPN2 acts as a key negative
regulator in T cells and contributes to central tolerance and the prevention of excessive responses to self-antigen in the periphery.1,2
The Janus kinases (JAK) 1 and 2 and the transcription factors STAT1, STAT3 and STAT5 are bona fide substrates of PTPN2.
Our studies indicate that global PTPN2 deficiency in mice enhances STAT5 signaling during early thymocyte development. DN
thymocytes isolated from Ptpn2–/– mice exhibited increased STAT5 phosphorylation at DN stages 2 and 3 resulting in elevated Bcl-2
levels and enhanced thymocyte survival. Studies with mixed bone marrow chimeras demonstrated that PTPN2-deficient thymocytes
outcompeted their wild type counterparts in lethally irradiated congenic wild type hosts at DN stages 3 and 4. In addition, T cell
progenitors isolated from the bone marrow of Ptpn2–/– mice transitioned faster through the DN-stages when compared to wild type
control cells using the OP9-DL1 system ex vivo. In contrast, retrovirally transduced over-expression of PTPN2 in bone-marrow chimeras
prevented progression beyond DN stage 2.
These results indicate that PTPN2 is required to restrain thymocyte selection at the DN stages in a cell-intrinsic manner through the
inhibition of the JAK/STAT5 pathway. These findings are consistent with studies showing that Interleukin-7 (IL-7)-induced STAT5
signaling is crucial for T cell development and survival and the progression of DN thymocytes to the double-positive (DP) stage. Our
results identify PTPN2 as a critical regulatory element in early T cell development.
1.
2.
Wiede et al. J Clin Invest. 2011 December 1; 121(12): 4758–4774.
Wiede et al. Nat.Commun. 2014 January; 5:3073.
352
The BTB-ZF transcriptional regulator, PLZF, modifies chromatin to restrain inflammatory cytokines
signaling programs
Anthony Sadler1, Fernando Rossello2, Stefan White1, Die Wang1, Michael Gantier1, Steve Gerondakis2, Bryan Williams1, Dakang
Xu
1. MIMR-PHI Institute of Medical Research, , Monash University, Clayton, Vic, Australia
2. Department of Clinical Haematology, Monash University, Melbourne, Vic, Australia
Inflammation is critical for host defense, but without appropriate control it can cause chronic disease, or even provoke fatal responses.
Here we identify a mechanism that limits the inflammatory response. Probing the responses of macrophages to the key sensory Toll-like
receptors, we identify that the BTB/POZ, zinc finger transcriptional regulator PLZF limits the expression of inflammatory cytokines
products. In accord with this, PLZF-deficient animals express higher levels of potent inflammatory cytokines and mount exaggerated
inflammatory responses to infectious stimuli. We show that this protective effect of PLZF is mediated via modulation of macrophage
activity. Temporal quantitation of inflammatory gene transcripts shows more rapid gene induction in the absence of PLZF. Genome-wide
analysis of histone modifications distinguish that PLZF establishes basal activity states of early response genes to maintain immune
homeostasis and limit damaging inflammation. We show that PLZF stabilizes a co-repressor complex that encompasses histone
deacetylase activity to control chromatin. Together with our previous demonstration that PLZF promotes the antiviral response, these
results suggest a strategy that could realize one of the major goals of immune therapy to retain immune resistance to pathogens while
curbing damaging inflammation.
353
Pronuclear injection of circular plasmids expressing hCas9 and gRNA generates mutant mice in
genes involved in immune responses.
Tomo Yonezawa1, Akihisa Oda1, Riho Kurata1, Sachiko Kubo1, Shyuhei Ogawa1, Yoichiro Iwakura1
1. Tokyo University of Science, Noda, CHIBA, Japan
Recently, the clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated (Cas) 9 system have been found
in some kind of bacteria’s defense system like a mammalian acquired immune system. This system develops as powerful tools to
modify the genome in many species including mammalian. In this system, gRNA recruits hCAS9 nuclease to the specific genomic locus
according to the base-pairing rule and then, introduces double strand breaks (DSBs). After the DSBs, non-homologous end-joining is
occurred and causes small insertion or deletion at the specific site. Thus, in order to rapidly obtain gene-deficient mouse in several
genes involved in immune responses, we designed over 10 gRNAs and validated their working with hCas9 expression in cultured cells.
After checking, we injected two plasmids expressing hCas9 and single gRNA, which is targeted at an inflammatory cytokine gene, into
the pronucleus of mouse zygote and transferred with pseudopregnant mice. Finally, we succeeded in getting several lines of the genedeficient mouse.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 127
354
Brain derived neurotrophic factor regulated immune responses in vivo
Yasuhiro Yoshida1, Yuan Song1, Kentaro Morita1
1. University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
It is well known that Brain derived neurotrophic factor (BDNF) is a one of neurotrophic factors and a key player for sustaining of
homeostasis in brain. On the contrary, there are few reports whether BDNF affects immune cells. We previously reported that BDNF
affects immune cells and induced regulatory T cells (Treg). In this study, we investigated whether BDNF has effects on immune cells in
vivo. At 24 hours after the BDNF administration by i.n., splenocytes were prepared. Flow cytometry demonstrated that BDNF
administration induced Foxp3 positive cells in splenocytes. To determine whether BDNF administration affected splenocytes,
proliferation of splenocytes were examined. The proliferation of splenocytes stimulated with mitogen (LPS) was attenuated. Additionally,
TNF-α production induced by LPS was lower than control in splenocytes. These results suggested that BDNF contributes immune
suppressive event in vivo.
355
Kupffer cell derived IL-1a promotes acetaminophen-induced liver injury
Chao Zhang1, Jin Feng1, Jun Du2, Shuo Yang1, Weihong Zhang3, Weihong Wang1, Hong Tang1
1. Institute of biophysics, Chinese Academy of Sciences, Chaoyang, China
2. The Institute of Biotechnology, Shanxi University, Taiyuan, China
3. Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
Cell necrosis can induce profound inflammation and potentially increases overall tissue injury. One important example is
acetaminophen-induced liver injury (AILI), in which the initial tissue injury caused by the toxicity of acetaminophen (APAP) is followed by
the second wave of tissue injury caused by cell necrosis and inflammatory responses. In the present study, we show that the TLR4dependent activation of IL-1a in kupffer cells is critical for the progression of AILI. Notably, interference of IL-1R1 or IL-1a, but not IL-1b
or NLRP3 inflammasome, exhibited reduced hepatic injury, as evidenced by decreased serum levels of alanine and aspartate
aminotransferases, decreased hepatic necrosis, decreased neutrophils and monocytes infiltration and improved survival of experimental
animals. The secreted IL-1a was derived from kupffer cells and the maturation and activation of IL-1a was dependent on TLR4/MyD88
signaling. Moreover, the infiltrated Gr-1 positive myeloid cells serve as target cells in response to IL-1a to contribute to liver injury.
Taken together, these findings indicate that inhibition of IL-1a signaling can alleviate the acute inflammatory responses and tissue injury,
and represent a potential therapeutic method in AILI.
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 128
Delegate Listing
Suad Abdirahman
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Olushola Oladapo Awojobi
Shoskilaos Ventures
Nigeria
[email protected]
Adrian Achuthan
University of Melbourne
Australia
[email protected]
Jeff Babon
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Mark Adams
Australian Biosearch
Australia
[email protected]
Yuriy Baglaenko
University of Toronto
Canada
[email protected]
Inna Afonina
Ghent University
Belgium
[email protected]
Paul Baker
Walter and Eliza Hall Institute
Australia
[email protected]
Afsar Ahmed
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Jesse Balic
MIMR-PHI Institute
Australia
[email protected]
Shizuo Akira
Osaka University
Japan
[email protected]
Aoi Akitsu
Tokyo University of Science
Japan
[email protected]
Andrei Alexeev
United States
[email protected]
Suhad AL-Yahya
KFSH&RC
Saudi Arabia
[email protected]
Corey Balinsky
National Institutes of Health
United States
[email protected]
Frances Balkwill
Barts Cancer Institute
United Kingdom
[email protected]
Marcin Baran
Trinity College Dublin
Ireland
[email protected]
Betsy Barnes
Rutgers Biomedical and Health
Sciences
United States
[email protected]
Thomas Angelovich
Burnet Institute
Australia
[email protected]
K. Mark Ansel
University of California
United States
[email protected]
Nikola Baschuk
La Trobe University
Australia
[email protected]
Engin Baturcam
University of Queensland
Australia
[email protected]
Ari Ari Waisman
Uni Mainz
Germany
[email protected]
Andy Bean
CSIRO
Australia
[email protected]
Juliana Ariffin
Institute for Molecular Bioscience
Australia
[email protected]
Irina Astrakhantseva
Lobachevsky State University of
Nizhniy Novgorod
Russia
[email protected]
Michael Beard
University of Adelaide
Australia
[email protected]
Sammy Bedoui
Doherty Institute for Infection &
Immunity
Australia
[email protected]
Gabrielle Belz
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Yinon Ben-Neriah
Hebrew University-Hadassah Medical
School
Israel
[email protected]
Cristina Bergamaschi
National Cancer Institute at Frederick
United States
[email protected]
Damien Bertheloot
University Hospital Bonn
Germany
[email protected]
Sonja Best
NIAID/NIH
United States
[email protected]
Kiran Bhaskar
University of New Mexico
United States
[email protected]
Subir Biswas
University of Calcutta
India
[email protected]
Kasia Blaszczyk
A. Mickiewicz University
Poland
[email protected]
Marie Bodinier
INRA
France
[email protected]
Sara Bonanzinga
Victorian Infectious Diseases
Reference Laboratory
Australia
[email protected]
Ernest Borden
Cleveland Clinic
United States
[email protected]
Katharina Borst
Twincore GmbH
Germany
[email protected]
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 129
Marieke Boshuizen
Academic Medical Center
Netherlands
[email protected]
Iain Campbell
University of Sydney
Australia
[email protected]
Gyeryung Choi
Naver
South Korea
[email protected]
Gregory Bouchaud
INRA
France
[email protected]
Ian Campbell
CSL Ltd
Australia
[email protected]
Amrita Chowdhury
Institute of Chemical Technology
India
[email protected]
Philippe Bouillet
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Zhihui (Helen) Cao
CSL Ltd
Australia
[email protected]
Anne-Marie Christensen
Queensland University of Technology
Australia
[email protected]
Natalia Castano Rodriguez
University of New South Wales
Australia
[email protected]
Tsung-Hsien Chuang
National Health Research Institutes
Taiwan
[email protected]
Virginia Castiglia
University of Vienna
Austria
[email protected]
Yeonseok Chung
Seoul National University
South Korea
[email protected]
Joseph Cavallari
McMaster University
Canada
[email protected]
Brian Clarke
SCMB, University of Queensland
Australia
[email protected]
Kenny Chan
Hospital for Sick Children
Canada
[email protected]
Daniel Clarke
Griffith University
Australia
[email protected]
Melody Chang
University of Melbourne
Australia
[email protected]
Rebecca Coll
Institute for Molecular Bioscience,
University of Queensland
Australia
[email protected]
Nollaig Bourke
Monash Institute of Medical Research
Australia
[email protected]
Karim J. Brandt
University of Geneva
Switzerland
[email protected]
Aaron Brice
University of Melbourne
Australia
[email protected]
Andrew Brooks
Institute for Molecular Bioscience,
University of Queensland
Australia
[email protected]
Gavin Brooks
Monash Institute of Medical Research
Australia
[email protected]
Sophie Broughton
St Vincent's Institute
Australia
[email protected]
Ross Chapman
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Stephanie Conos
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Alison Browning
Monash University
Australia
[email protected]
Stephane Chappaz
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Bruno Conti
UNESP
Brazil
[email protected]
Annie Bruns
Northwestern University
United States
[email protected]
Ya-Lei Chen
National Kaoshiung Normal University
Taiwan
[email protected]
Andrew Cook
University of Melbourne
Australia
[email protected]
Vipin Bulani
Institute of Chemical Technology
India
[email protected]
Zhijian 'James' Chen
University of Texas Southwestern
Medical Center
United States
[email protected]
Matthew Cooper
University of Queensland
Australia
[email protected]
Sebastien Burel
ISIS Pharmaceuticals
United States
[email protected]
Samantha Busfield
Nexvet
Australia
[email protected]
Irina Caminschi
Burnet Institute
Australia
[email protected]
Choy-Hoong Chew
Faculty of Science, Universiti Tunku
Abdul Rahman
Malaysia
[email protected]
Steven Cho
MIMR - The Ritchie Centre
Australia
[email protected]
Mariana Corrales Benitez
Monash University
Australia
[email protected]
Ben Cotton
University of Melbourne
Australia
[email protected]
Daniel Croker
University of Queensland
Australia
[email protected]
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 130
Daniel Cua
Merck
United States
[email protected]
Poshmaal Dhar
University of Melbourne
Australia
[email protected]
Mark Febbraio
Baker IDI Heart & Diabetes Institute
Australia
[email protected]
Yanfang Cui
Monash university
Australia
[email protected]
Evdokia Dimitriadis
Prince Henry's Institute of Medical
Research
Australia
[email protected]
Zoltan Fehervari
Nature Publishing Group
United Kingdom
[email protected]
Helen Cumming
MIMR-PHI
Australia
[email protected]
Tyler Curiel
UT Health Science Center
United States
[email protected]
Claudia Dominguez
Yale University
United States
[email protected]
Jennifer Dowling
MIMR-PHI Institute
Australia
[email protected]
Dhouha Daassi
Tsukuba University
Japan
[email protected]
Alec Drew
Nexvet
Australia
[email protected]
Laura Dagley
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Aïcha Daher
Lady Davis Institute for Medical
Research
Canada
[email protected]
Joan Durbin
Rutgers - New Jersey Medical School
United States
[email protected]
Russell Durbin
Rutgers - New Jersey Medical School
United States
[email protected]
Scott Durum
National Cancer Institute
United States
[email protected]
Vinh Dao
University of Texas
United States
[email protected]
Sophia Davidson
National Institute for Medical Research
United Kingdom
[email protected]
Sarah Edwards
Trinity College Dublin
Ireland
[email protected]
Colleen Elso
St Vincent's Institute
Australia
[email protected]
Hugh Davis
Janssen R&D, LLC
United States
[email protected]
Kimberley D'Costa
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Dominic De Nardo
Institute of Innate Immunity
Germany
[email protected]
Seong Kug Eo
College Of Veterinary Medicine
South Korea
[email protected]
Matthias Ernst
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Nicole De Weerd
Monash Institute of Medical Research
Australia
[email protected]
Nima Etemadi
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Padmini Deshpande
Institute of Chemical Technology
India
[email protected]
Inês F. Amado
Pasteur Institute
France
[email protected]
Walter Ferlin
NovImmune S.A
Switzerland
[email protected]
Daniel Fernandez Ruiz
Peter Doherty Institute
Australia
[email protected]
Antonio Ferrante
SA Pathology at Women's and
Children's Hospital
Australia
[email protected]
Manuel Ferreira
Queensland Institute of Medical
Research
Australia
[email protected]
Richard Ferrero
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Rosemary French
Monash University
Australia
[email protected]
Conor Finlay
Trinity College Dublin
Ireland
[email protected]
Eleanor Fish
University Health Network
Canada
[email protected]
Kate Fitzgerald
University of Massachusetts Medical
School
United States
[email protected]
Patricia Fitzgerald-Bocarsly
Rutgers - New Jersey Medical School
United States
[email protected]
Andrew Fleetwood
University of Melbourne
Australia
[email protected]
Niels Foged
Euro Diagnostica AB
Sweden
[email protected]
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 131
Sam Forster
MIMR-PHI Insitute of Medical Research
Australia
[email protected]
Nathalie Grandvaux
CRCHUM/Université de Montréal
Canada
[email protected]
James Harris
Monash University (MIMR)
Australia
[email protected]
Kathryn Friend
Australian Biosearch
Australia
[email protected]
Michael Griffin
University of Melbourne
Australia
[email protected]
Angela Harrison
Australia
[email protected]
Svenja Fritzlar
University of Melbourne
Australia
[email protected]
Ewelina Grywalska
Medical University of Lublin
Poland
[email protected]
Ka Yee Fung
University of Melbourne
Australia
[email protected]
Raffi Gugasyan
Burnet Institute
Australia
[email protected]
Cem Gabay
University of Geneva
Switzerland
[email protected]
Audrey Gustin
University of Luxembourg
Luxembourg
[email protected]
Hans Henrik Gad
Aarhus University
Denmark
[email protected]
Reena Halai
University of Queensland
Australia
[email protected]
Michael Gantier
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Otto Haller
University of Freiburg
Switzerland
[email protected]
David Gearing
Nexvet
Australia
[email protected]
Carolyn Geczy
University of New South Wales
Australia
[email protected]
Jason Gill
TEVA Biologics
Australia
[email protected]
Florent Ginhoux
Agency for Science, Technology and
Research (A*STAR)
Singapore
[email protected]
Gabrielle Goldberg
Walter and Eliza Hall Institute
Australia
[email protected]
Daniel Gough
Monash Institute of Medical Research
Australia
[email protected]
Jodee Gould
MIMR-PHI Institute
Australia
[email protected]
John Hamilton
University of Melbourne
Australia
[email protected]
Chaofeng Han
Institute of Immunology, Second
Military Medical University
China
[email protected]
Mi-Soon Han
Severance Hospital, Yonsei University
College of Medicine
South Korea
[email protected]
Sang-Chul Han
Jeju National University
South Korea
[email protected]
Moritz Haneklaus
Trinity College Dublin
Ireland
[email protected]
Ashraful Haque
QIMR Berghofer
Australia
[email protected]
Daniel Harari
Weizmann Institute of Science
Israel
[email protected]
Sharni Hardcastle
Griffith University
Australia
[email protected]
Elizabeth Hartland
University of Melbourne
Australia
[email protected]
Dallas Hartman
Nexvet
Australia
[email protected]
Rune Hartmann
Aarhus University
Denmark
[email protected]
Mark Hedger
Monash Institute of Medical Research,
Prince Henry's Institute
Australia
[email protected]
Karla Helbig
Australia
[email protected]
Tim Hercus
Centre for Cancer Biology
Australia
[email protected]
Karl Herron
Abcam Australia
Australia
[email protected]
Paul Hertzog
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Margaret Hibbs
Monash University
Australia
[email protected]
Yuka Hiroshima
University of New South Wales
Australia
[email protected]
Jeremy Hirota
University of British Columbia
Canada
[email protected]
Kathryn Hjerrild
Nexvet
Australia
[email protected]
Hubertus Hochrein
Bavarian Nordic
Germany
[email protected]
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 132
Markus Hofer
University of Sydney
Australia
[email protected]
Stacy Horner
Duke University Medical Center
United States
[email protected]
Veit Hornung
University Hospital, University of Bonn
Germany
[email protected]
Alan Hsu
University of Newcastle
Australia
[email protected]
Kenneth Hsu
University New South Wales
Australia
[email protected]
Amanda Huber
University of Michigan
United States
[email protected]
Anissa Jabbour
Walter & Eliza Hall Institute of Medical
Research
Australia
[email protected]
Brendan Jenkins
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Nicholas Huntington
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Farida Hussan
Universiti Kebangsaan Malaysia
Malaysia
[email protected]
Melanie Hutton
Monash University
Australia
[email protected]
Soo-Seok Hwang
Life-Science Department of Sogang
University
South Korea
[email protected]
Juliana I. Hori
FMRP/USP
Brazil
[email protected]
Aaron Irving
Duke-NUS Graduate Medical School,
Singapore
Singapore
[email protected]
Yoichiro Iwakura
Tokyo University of Science
Japan
[email protected]
Alexander Karnowski
CSL Ltd
Australia
[email protected]
Indzi Katik
Australian Biosearch
Australia
[email protected]
Shaoquan Ji
BioLegend
United States
[email protected]
Hiroki Kato
Japan
[email protected]
Zhengfan Jiang
Peking University
China
[email protected]
Naomi Kawaguchi
University of New South Wales
Australia
[email protected]
Simon Jones
Medical School, Cardiff University
United Kingdom
[email protected]
Emma Kay
William Harvey Research Institute,
QMUL
United Kingdom
[email protected]
Haiyoung Jung
KRIBB
South Korea
[email protected]
Ian Humphreys
Cardiff University
United Kingdom
[email protected]
Australia
[email protected]
So Ri Jung
University of Sydney
Australia
[email protected]
Young Su Jung
Sungkyunkwan university
South Korea
[email protected]
Nadeem Kaakoush
University of New South Wales
Australia
[email protected]
Dhan Kalvakolanu
University of Maryland
United States
[email protected]
Tomonori Kamiya
Research Institute for Biomedical
Sciences, Tokyo University of Science
Japan
[email protected]
Winnie Kan
Centre for Cancer Biology
Australia
[email protected]
Thirumala-Devi Kanneganti
St Jude Children's Research Hospital
United States
[email protected]
Cheng-Yuan Kao
National Health Research Institutes
Taiwan
[email protected]
Ronan Kapetanovic
University Of Queensland
Lukasz Kedzierski
Walter & Eliza Hall Institute of Medical
Research
Australia
[email protected]
Michael Keir
Lonza
Australia
[email protected]
Khalid S. A. Khabar
King Faisal Specialist Hospital and
Research Centre
Saudi Arabia
[email protected]
Adnan Khan
Trinity Biomedical Sciences Institute,
Trinity College Dublin
Ireland
[email protected]
Parvez Khan
Centre for Interdisciplinary Research in
Basic Sciences
India
[email protected]
Hsu Wei Khiew
University Of Melbourne
Australia
[email protected]
Dan Kho
The University of Auckland
New Zealand
[email protected]
Katrina Ki
Australian Red Cross Blood Service
Australia
[email protected]
Benjamin Kile
Walter and Eliza Hall Institute of
Medical Research
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 133
Australia
[email protected]
Sung-Jo Kim
Pusan National University School of
Dentistry
South Korea
[email protected]
Elodie Kip
WIV-ISP
Belgium
[email protected]
Mireille Lahoud
Burnet Institute
Australia
[email protected]
Christophe Lamaze
Institut Curie
France
[email protected]
Tali Lang
Monash University Australia
Australia
[email protected]
Tatiana Kolesnik
Walter & Eliza Hall Institute of Medical
Research
Australia
[email protected]
Iryna Kolosenko
Karolinska Institutet
Sweden
[email protected]
Tomasz Kordula
Virginia Commonwealth University
United States
[email protected]
Sergei Kotenko
Rutgers, New Jersey Medical School
United States
[email protected]
Pankaj Kothavade
Institute of chemical Technology
India
[email protected]
Shruti Krishnan
University of Western Australia
Australia
[email protected]
Hyung-Keun Ku
Korea Yakult
South Korea
[email protected]
Yoshinao Kubo
Nagasaki University
Japan
[email protected]
Marcel Kuhny
University Clinic RWTH AACHEN
Germany
[email protected]
Florian Kurschus
University Medical Center of the
Johannes Gutenberg-University
Germany
[email protected]
Elina L Zuniga
University of California
United States
[email protected]
Derek Lacey
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Australia
[email protected]
David Levy
NYU School of Medicine
United States
[email protected]
Maria Liaskos
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Katherine Langley
Monash University
Australia
[email protected]
Nicholas Liau
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Chun Wang Jason Lao
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Dawn Lin
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Andrew Larner
Virginia Commonwealth University
United States
[email protected]
Helene Minyi Liu
National Taiwan University
Taiwan
[email protected]
Eicke Latz
University of Bonn
Germany
[email protected]
Xiao Liu
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Kate Lawlor
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Mathieu Le Gars
Institut Pasteur
France
[email protected]
Jun-young Lee
Kon-yang University
South Korea
[email protected]
Ming-Chin Lee
University of Melbourne
Australia
[email protected]
Sang-Myeong Lee
Chonbuk National Universrity
South Korea
[email protected]
Seojin Lee
Naver
South Korea
[email protected]
Joachim Lehmann
Novartis Institutes for Biomedical
Research
Switzerland
[email protected]
Yew Ann Leong
Monash University
Yiliu Liu
Canada
[email protected]
Zhi Liu
University of North Carolina
United States
[email protected]
Liza Ljungberg
Sweden
[email protected]
Camden Lo
MIMR-PHI Institute
Australia
[email protected]
Zhixuan Loh
University Of Queensland
Australia
[email protected]
Carlin Long
Denver Health Medical Center
United States
[email protected]
Yueh-Ming Loo
University of Washington
United States
[email protected]
Angel Lopez
Centre for Cancer Biology
Australia
[email protected]
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 134
Cynthia Louis
University of Melbourne
Australia
[email protected]
Ashley Mansell
Monash Institute of Medical Research
Australia
[email protected]
Jun Ting Low
Walter & Eliza Hall Institute of Medical
Research
Australia
[email protected]
Isabelle Marié
NYU School of Medicine
United States
[email protected]
Zoe Marks
MIMR-PHI Institute
Australia
[email protected]
Pei Ching (Regine) Low
UCL Cancer Institute
United Kingdom
[email protected]
Tao Lu
Indiana University
United States
[email protected]
Seth Masters
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Erik Lundgren
Umeå University
Sweden
[email protected]
Mariko Matsui
Institut Pasteur de Nouvelle-Calédonie
New Caledonia
[email protected]
Kelli MacDonald
QIMR Berghofer
Australia
[email protected]
Kate McArthur
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Fabienne Mackay
Monash University, Central Clinic
School
Australia
[email protected]
Brent Mckenzie
Genentech
United States
[email protected]
Raquel Magalhaes
eBioscience, an Affymetrix company
Singapore
[email protected]
Louise McLeod
MIMR-PHI Institute
Australia
[email protected]
Tandeka Magcwebeba
Stellenbosch University
South Africa
[email protected]
Mualla Mcmanus
University of Sydney
Australia
[email protected]
Tanel Mahlakoiv
University of Freiburg Medical Center
Germany
[email protected]
Carina Melo
University of São Paulo
Brazil
[email protected]
Saeed Mahmoudi
Deakin University
Australia
[email protected]
Ellen Menkhorst
MIMR-PHI Institute
Australia
[email protected]
Pilvi Maliniemi
University of Helsinki
Finland
[email protected]
Lisa Mielke
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Si Ming Man
St. Jude Children's Research Hospital
United States
[email protected]
Niamh Mangan
MIMR-PHI
Australia
[email protected]
Offir Manor
PeproTech
Australia
[email protected]
Alison Mildenhall
Walter and Eliza Hall Institute
Australia
[email protected]
Steven Mileto
Monash University
Australia
[email protected]
Kingston Mills
Trinity Biomedical Sciences Institute
Ireland
[email protected]
Hazel Mitchell
University of New South Wales
Australia
[email protected]
Denise Monack
Stanford University Stanford School of
Medicine
United States
[email protected]
Katherine Monaghan
CSL Limited
Australia
[email protected]
Alessandra Mortellaro
Singapore Immunology Network (SIgN)
Singapore
[email protected]
Gregory Moseley
University of Melbourne
Australia
[email protected]
Richard Müller-Brunotte
Karolinska Institutet
Sweden
[email protected]
Usma Munawara
SA Pathology
Australia
[email protected]
Shashirekha Mundhra
Weill Cornell Graduate School of
Medical Sciences
United States
[email protected]
Masaaki Murakami
Hokkaido University
Japan
[email protected]
Andrew Murphy
Baker IDI Heart and Diabetes Institute
Australia
[email protected]
Nontobeko Mvubu
University of KwaZulu-Natal
South Africa
[email protected]
Thomas Naderer
Monash University
Australia
[email protected]
Jae-Hwan Nam
Catholic University Of Korea
South Korea
[email protected]
Garrett Ng
Murdoch Childrens Research Institute
Australia
[email protected]
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 135
Ivan Ng
Monash University
Australia
[email protected]
Luke O'Neill
Trinity College Dublin
Ireland
[email protected]
Paul Nguyen
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Lorraine O'Reilly
Walter & Eliza Hall Institute of Medical
Research
Australia
[email protected]
Tan Nguyen
Walter and Eliza Hall Institute
Australia
[email protected]
Ken Pang
Walter and Eliza Hall Institute
Australia
[email protected]
Thi Loan Anh Nguyen
VIB-Autoimmune Genetics
Belgium
[email protected]
Geena Paramel
Orebro University, Department of health
and Medical science
Sweden
[email protected]
Sandra Nicholson
Walter & Eliza Hall Institute of Medical
Research
Australia
[email protected]
Hye-Lim Park
Catholic University Of Korea
South Korea
[email protected]
Nicos Nicola
Walter and Eliza Hall Institute
Australia
[email protected]
Jong-Hwan Park
Konyang University
South Korea
[email protected]
Claudia Nold
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Belinda Parker
La Trobe University
Australia
[email protected]
Marcel Nold
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Mizuho Nosaka
Wakayama Medical University
Japan
[email protected]
Gabriel Nunez
University of Michigan Medical School
United States
[email protected]
Roza Nurieva
MD Anderson Cancer Center
United States
[email protected]
Robert O'Donoghue
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Meredith O'Keeffe
Burnet Institute
Australia
[email protected]
Olusegun Onabajo
National Institutes of Health (NIH)
United States
[email protected]
Matthias Parrini
University of Veterinary Medicine
Austria
[email protected]
Barry Parsons
Ballinger Family Medical Practise
Australia
[email protected]
Suzanne Paz
Isis Pharmaceuticals
United States
[email protected]
Australia
[email protected]
Anita Pinar
MIMR-PHI Institute of Medical Resarch
Australia
[email protected]
Ashleigh Poh
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Ludmila Prokunina-Olsson
National Cancer Institute, NIH
United States
[email protected]
Tracy Putoczki
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Kylie Quinn
Department of Microbiology and
Immunology, University of Melbourne
Australia
[email protected]
Rajes Qvist
university of Malaya
Malaysia
[email protected]
Sukanya Raghavan
University of Gothenburg
Sweden
[email protected]
Steve Ralph
Genvax Pty Ltd
Australia
[email protected]
Hilario Ramos
Gilead Sciences
United States
[email protected]
Jaclyn Pearson
University of Melbourne
Australia
[email protected]
Maryam Rashidi
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Karin Pelka
University of Bonn
Germany
[email protected]
Mahesh Raundhal
University of Pittsburgh
United States
[email protected]
Leesa Pennell
University of Toronto and University
Health Network
Canada
[email protected]
Jai Rautela
LaTrobe Institute for Molecular
Sciences & Peter MacCallum Cancer
Centre
Australia
[email protected]
Cathleen Pfefferkorn
University Medical Center Freiburg
Germany
[email protected]
Mireia Recasens Torné
University Autonomous of Barcelona
Spain
[email protected]
Simon Phipps
University of Queensland
Jay Reddy
University of Nebraska
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 136
United States
[email protected]
Marvin Sandoval
NYU School of Medicine
United States
[email protected]
Srinivasan Rengachari
EMBL
France
[email protected]
Joseph Reynolds
Rosalind Franklin University
United States
[email protected]
Natalia Rinis
University Clinic RWTH Aachen
Germany
[email protected]
Fabiana Rizzo
Istituto Superiore di Sanità
Italy
[email protected]
Avril Robertson
University of Queensland
Australia
[email protected]
Stefan Rose-John
University of Kiel
Germany
[email protected]
Tony Rowe
CSL Ltd
Australia
[email protected]
Ina Rudloff
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Saleela Ruwanpura
MIMR-PHI Institute of Medical Resarch
Australia
[email protected]
Catarina Sacristan
Rockefeller University Press
United States
[email protected]
Muhammad Saeed
Murdoch childrens Research Institute
Australia
[email protected]
Karina Santiago
UNESP
Brazil
[email protected]
Soroush Sarvestani
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Karen Scalzo-Inguanti
CSL Ltd
Australia
[email protected]
William Schneider
Rockefeller University
United States
[email protected]
Glen Scholz
University of Melbourne
Australia
[email protected]
Kate Schroder
University of QLD
Australia
[email protected]
Margret Schuller
PeproTech
Australia
[email protected]
YunJi Seo
Kyung Hee University
South Korea
[email protected]
Feng Shao
National Institute of Biological Sciences
China
[email protected]
Smriti Sharma
Insititute of Medical Sciences, Banaras
Hindu University
India
[email protected]
Vitaliya Sagulenko
University of Queensland
Australia
[email protected]
Kenji Shimizu
Center for Animal Disease Models,
Research Institute for Biomedical
Sciences, Tokyo University of Science
Japan
[email protected]
Reem Saleh
University of Melbourne
Australia
[email protected]
Kalpana Shrivastava
Universidad Autonoma de Barcelona
Spain
[email protected]
Maite Sanchez-Aparicio
ICAHN SCHOOL OF MEDICINE AT
MOUNT SINAI
United States
[email protected]
Anabel Silva
CSL Limited
Australia
[email protected]
United States
[email protected]
Allan Sirsjö
Örebro university
Sweden
[email protected]
Martine Smale
Transnetyx
United States
[email protected]
Sara Small
University of Pennsylvania
United States
[email protected]
Ida Smith
Chr. Hansen
Denmark
[email protected]
Rebecca Smith
Victorian Infection and Immunity
Network Industry Alliance
Australia
[email protected]
Mark Smyth
QIMR Berghofer Medical Research
Institute
Australia
[email protected]
Mark Smythe
Protagonist Therapeutics
Australia
[email protected]
Vicki Sorto
CSL Limited
Australia
[email protected]
Julia Spanier
Twincore
Germany
[email protected]
Helmut Sparrer
Novartis
Switzerland
[email protected]
Igor Splichal
Institute of Microbiology, ASCR
Czech Republic
[email protected]
Alla Splichalova
Institute of Microbiology, ASCR
Czech Republic
[email protected]
Kate Stacey
University of Queensland
Australia
[email protected]
Peter Staeheli
University Medical Center Freiburg
Germany
[email protected]
Robert Silverman
Cleveland Clinic
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 137
George Stark
Cleveland clinic
United States
[email protected]
Australia
[email protected]
Malcolm Starkey
University of Newcastle Australia
Australia
[email protected]
Jennifer Stow
University of Queensland
Australia
[email protected]
Elizabeth Thatcher
U Mass Medical School
United States
[email protected]
Belinda Thomas
MIMR-PHI Institute
Australia
[email protected]
Sara Thygesen
SCMB
Australia
[email protected]
William Strohl
Janssen R&D
United States
[email protected]
James Stunden
Institute for Innate Immunity
Germany
[email protected]
Andrea Stutz
University of Bonn
Germany
[email protected]
Matt Sweet
University of Queensland
Australia
[email protected]
Taylor Syme
University of Sydney
Australia
[email protected]
Hong Tang
Chinese Academy of Sciences
China
[email protected]
Maria Tanzer
Walter & Eliza Hall Institute of Medical
Research
Australia
[email protected]
Mohd Tarique
All India Institute of Medical Sciences
(AIIMS)
India
[email protected]
Michelle Tate
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Jan Tavernier
UGent/VIB
Belgium
[email protected]
Nicodemus Tedla
University New South Wales
Australia
[email protected]
Michele Teng
QIMR Berghofer Medical Research
Institute
Rey Tiquia
University of Melbourne
Australia
[email protected]
Michael Tovey
CNRS
France
[email protected]
Shota Toyoshima
Tokyo University of Science
Japan
[email protected]
Nick Tsichlis
Lonza
Australia
[email protected]
Denis Tvorogov
Center for Cancer Biology
Australia
[email protected]
Jason Twohig
Cardiff University
United Kingdom
[email protected]
Hazel Tye
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Jasim Uddin
Queensland University
Australia
[email protected]
Dale T Umetsu
Genentech
United States
[email protected]
Kazuko Uno
Louis Pasteur Center for Medical
Research
Japan
[email protected]
Gino Vairo
CSL Limited
Australia
[email protected]
Parimala Vajjhala
University of Queensland
Australia
[email protected]
Renee van der Sluis
Doherty Institute, University of
Melbourne
Australia
[email protected]
Antiopi Varelias
QIMR Berghofer Medical Research
Institute
Australia
[email protected]
Lauren Vaughn
University of South Carolina
United States
[email protected]
Anne Verhagen
CSL
Australia
[email protected]
Elena Vigano
Agency for Science, Technology and
Research (A*STAR)
Singapore
[email protected]
Swarna Lekha Vijayaraj
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Nàdia Villacampa
University Autonomous of Barcelona
Spain
[email protected]
Jose Villadangos
University of Melbourne
Australia
[email protected]
James Vince
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Uwe Vinkemeier
University of Nottingham
United Kingdom
[email protected]
Carola Vinuesa
John Curtin School of Medical
Research, The Australian National
University
Australia
[email protected]
Nazarii Vitak
SCMB, University of Queensland
Australia
[email protected]
Matthias von Gamm
Helmholtz Zentrum München
Germany
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 138
[email protected]
Anna Walduck
RMIT University
Australia
[email protected]
Adam Wall
University of Queensland
Australia
[email protected]
Shih-Min Wang
National Cheng Kung University and
Hospital,
Taiwan
[email protected]
Xiao-Fan Wang
Duke University Medical Center
United States
[email protected]
Peter Wark
Hunter Medical Research Institute
Australia
[email protected]
Kevin Watanabe-Smith
Oregon Health and Science University
United States
[email protected]
Wolfgang Weninger
Centenary Institute
Australia
[email protected]
Rhiannon Werder
University of Queensland
Australia
[email protected]
Michael White
Walter and Eliza Hall Institute of
Medical Research
Australia
[email protected]
Paul Whitney
Doherty Institute
Australia
[email protected]
Ian Wicks
Walter and Eliza Hall Institute
Australia
[email protected]
Florian Wiede
Monash University
Australia
[email protected]
Nicholas Wilson
CSL Limited
Australia
[email protected]
Hiroki Yoshida
Saga University, Faculty of Medicine
Japan
[email protected]
Anders Woetmann
University of Copenhagen
Denmark
[email protected]
Yasuhiro Yoshida
University of Occupational and
Environmental Health, Japan
Japan
[email protected]
Naomi Wong
National Centre Of Neuroimmunology
And Emerging Diseases
Australia
[email protected]
Howard Young
National Cancer Institute at Frederick
United States
[email protected]
Craig Wright
Deakin University
Australia
[email protected]
Di Yu
Monash University
Australia
[email protected]
Jim Xiang
University of Saskatchewan
Canada
[email protected]
Hua Yu
Beckman Research Institute at City of
Hope Comprehensive Cancer Center
United States
[email protected]
Hui Xiao
Institut Pasteur of Shanghai
China
[email protected]
Dakang Xu
Monash University
Australia
[email protected]
Meilang Xue
University of Sydney
Australia
[email protected]
Min Yi
Hokkaido University
Japan
[email protected]
Howard Yim
MIMR-PHI Institute of Medical
Research
Australia
[email protected]
Liang Yu
Monash Institute of Medical Research
Australia
[email protected]
Alina Zamoshnikova
University of Queensland
Australia
[email protected]
Chao Zhang
Institute of biophysics, Chinese
Academy of Sciences
China
[email protected]
Kangjian Zhang
Chinese Academy Of Sciences
China
[email protected]
Yongliang Zhang
National University of Singapore
Singapore
[email protected]
Dan Yin
Huazhong University of Science and
Technology
China
[email protected]
YongGang Zhao
GSK
China
[email protected]
Tomo Yonezawa
Tokyo University of Science
Japan
[email protected]
Xiaohui Zhou
Institute of Heart Failure
China
[email protected]
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 139
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 140
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 141
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 142
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 143
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 144
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 145
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 146
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 147
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 148
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 149
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 150
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 151
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 152
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 153
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 154
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 155
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 156
Notes
The International Cytokine and Interferon Society 2014 Annual Meeting - Page 157